Language selection

Search

Patent 2325597 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2325597
(54) English Title: MULTIPARAMETER FACS ASSAYS TO DETECT ALTERATIONS IN CELLULAR PARAMETERS AND TO SCREEN SMALL MOLECULE LIBRARIES
(54) French Title: DOSAGES A PARAMETRES MULTIPLES POUR TRIEURS DE CELLULES PAR FLUORESCENCE SERVANT A DETECTER DES MODIFICATIONS DE PARAMETRES CELLULAIRES ET A CRIBLER DES BIBLIOTHEQUES DE PETITES MOLECULES
Status: Expired
Bibliographic Data
(51) International Patent Classification (IPC):
  • G01N 33/50 (2006.01)
  • G01N 33/533 (2006.01)
  • G01N 33/569 (2006.01)
(72) Inventors :
  • FISHER, JOSEPH (United States of America)
  • LORENS, JAMES (United States of America)
  • PAYAN, DONALD (United States of America)
  • ROSSI, ALEXANDER (United States of America)
(73) Owners :
  • RIGEL PHARMACEUTICALS, INC. (United States of America)
(71) Applicants :
  • RIGEL PHARMACEUTICALS, INC. (United States of America)
(74) Agent: SMART & BIGGAR
(74) Associate agent:
(45) Issued: 2010-09-21
(86) PCT Filing Date: 1999-04-16
(87) Open to Public Inspection: 1999-10-28
Examination requested: 2004-04-13
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US1999/008345
(87) International Publication Number: WO1999/054494
(85) National Entry: 2000-10-16

(30) Application Priority Data:
Application No. Country/Territory Date
09/062,330 United States of America 1998-04-17
09/157,748 United States of America 1998-09-21

Abstracts

English Abstract

The invention relates to novel methods of detecting alterations in cell cycle regulation in a cell or a cell population and screening for agents capable of modulating cell cycle regulation through the use of multiparameter assays and a fluorescence-activated cell sorter (FACS) machine.


French Abstract

Dans la présente invention, on divulgue de nouvelles méthodes de détection des modifications de la régulation du cycle cellulaire d'une cellule ou d'une population de cellules et de criblage d'agents pouvant moduler la régulation du cycle cellulaire, au moyen d'épreuves multiparamétriques et d'un appareil de triage de cellules activé par fluorescence.

Claims

Note: Claims are shown in the official language in which they were submitted.




CLAIMS:

1. A method comprising:

a) introducing a nucleic acid that encodes a
candidate bioactive agent into a population of cells to
produce contacted cells that produce said candidate
bioactive agent; and

b) sorting said contacted cells on the basis of at
least five cellular parameters using a fluorescence-
activated cell sorter (FACS), and

c) identifying a cell having a phenotype that is
altered by said candidate bioactive agent.


2. A method according to claim 1 wherein a library of
nucleic acids, each said nucleic acid encoding a bioactive
agent, is introduced into said population.


3. A method of screening for a bioactive agent
capable of altering a cellular phenotype, said method
comprising:

a) introducing a library of nucleic acids, each
nucleic acid in said library encoding a candidate bioactive
agent, into a population of cells; and

b) sorting said cells in a FACS machine by
separating said cells on the basis of at least five cellular
parameters.


4. A method according to claim 3 wherein said library
is a retroviral library.


5. A method according to claim 3 or 4 wherein said
cellular phenotype is exocytosis and said cellular
parameters are selected from the group consisting of light


56



scattering, fluorescent dye uptake, fluorescent dye release,
annexin granule binding, surface granule enzyme activity,
and quantity of granule specific proteins.


6. A method according to claim 5 further comprising
subjecting said cells to conditions that cause exocytosis in
cells in which said library of nucleic acids has not been
introduced.


7. A method according to claim 3 or 4 wherein said
cellular phenotype is cell cycle regulation and said
cellular parameters comprise cell viability, proliferation,
and cell phase.


8. A method according to claim 3, 4, 5, 6 or 7
wherein said nucleic acids encode fusion proteins
comprising:

a) said candidate bioactive agents; and
b) a detectable moiety.


9. A method according to claim 1, 2, 3, 4, 5, 6, 7
or 8 wherein said cells are tumor cells.


10. A method according to claim 8 wherein said
detectable moiety is a fluorescent protein.


57

Description

Note: Descriptions are shown in the official language in which they were submitted.



CA 02325597 2008-09-10
,77245-54

MULTIPARAMETER FACS ASSAYS TO DETECT
ALTERATIONS IN CELLULAR PARAMETERS AND TO SCREEN SMALL MOLECULE LIBRARIES
FIELD OF THE INVENTION
The invention relates to novel methods of detecting alterations in cellular
parameters, and particularly
for screening libraries of small molecules such as combinatorial chemical
libraries of organic
molecules, including peptides and other chemical libraries, for binding to
target molecules, using
fluoroscence-activated cell sorting (FACS) machines.
BACKGROUND OF THE INVENTION

The field of drug discovery and screening of drug candidates to identify lead
compounds is rapidly
expanding. Traditional approaches to identify and characterize new and useful
drug candidates
include the isolation of natural products or synthetic preparation, followed
by testing against either
known or unknown targets. See for example WO 94/24314, Gallop et al., J. Med.
Chem. 37(9):1233
(1994); Gallop et al., J. Med. Chem. 37(10):1385 (1994); Eliman, Acc. Chem.
Res. 29:132 (1996);
Gordon et al., E. J. Med. Chem. 30:388s (1994); Gordon et al., Acc. Chem. Res.
29:144 (1996); WO
95/12608.
The screening of these libraries is done in a variety of ways. One approach
involves attachment to
beads and visualization with dyes; see Neslter et al., Bioorg. Med. Chem.
Lett. 6(12):1327 (1996).
Another approach has utilized beads and fluorescence activated cell sorting
(FACS); see Needles et
al., PNAS USA 90:10700 (1993), and Vetter et al., Bioconjugate Chem. 6:319
(1995).
Fluorescence activated cell sorting (FACS), also called flow cytometry, is
used to sort individual cells
on the basis of optical properties, including fluorescence. It is generally
fast, and can result in
screening large populations of cells in a relatively short period of time.

1


CA 02325597 2000-10-16

WO 99/54494 PCTIUS99/08345
There are a number of instances where rapid and inexpensive screens such as
FACS screens would
be of particular interest. On such area is in cell cycle assays. Cells cycle
through various stages of
growth, starting with the M phase, where mitosis and cytoplasmic division
(cytokinesis) occurs. The M
phase is followed by the G1 phase, in which the cells resume a high rate of
biosynthesis and growth.
The S phase begins with DNA synthesis, and ends when the DNA content of the
nucleus has
doubled. The cell then enters G2 phase, which ends when mitosis starts,
signaled by the appearance
of condensed chromosomes. Terminally differentiated cells are arrested in the
GI phase, and no
longer undergo cell division.
The hallmark of a malignant cell is uncontrolled proliferation. This phenotype
is acquired through the
accumulation of gene mutations, the majority of which promote passage through
the cell cycle. Cancer
cells ignore growth regulatory signals and remain committed to cell division.
Classic oncogenes, such
as ras, lead to inappropriate transition from G1 to S phase of the cell cycle,
mimicking proliferative
extracellular signals. Cell cycle checkpoint controls ensure faithful
replication and segregation of the
genome. The loss of cell cycle checkpoint control results in genomic
instability, greatly accelerating the
accumulation of mutations which drive malignant transformation. Hence,
checkpoint regulators, such
as p53 and ATM (ataxia telangiectasia mutated), also function as tumor
suppressors. Thus,
modulating cell cycle checkpoint pathways with therapeutic agents could
exploit the differences
between normal and tumor cells, both improving the selectivity of radio- and
chemotherapy, and
leading to novel cancer treatments.

Accordingly, it is an object of the invention to provide compositions and
methods useful in screening
for modulators of cell cycle checkpoint regulation.
Another area for which rapid screening methods would find particular use is in
the area of assays of
exocytosis. Exocytosis is the fusion of secretory vesicles with the cellular
plasma membrane, and has
two main functions. One is the discharge of the vesicle contents into the
extracellular space, and the
second is the incorporation of new proteins and lipids into the plasma
membrane itself.
Exocytosis can be divided into two classes: constitutive and regulated. All
eukaryotic cells exhibit
constitutive exocytosis, which is marked by the continuous fusion of the
secretory vesicles after
formation. Regulated exocytosis is restricted to certain cells, including
exocrine, endocrine and
neuronal cells. Regulated exocytosis results in the accumulation of the
secretory vesicles that fuse
with the plasma membrane only upon receipt of an appropriate signal, usually
(but not always) an
increase in the cytosolic free Ca2+ concentration.

2


CA 02325597 2008-09-10
77245-54

Regulated exocytosis is crucial to many specialized cells, and often a
particular cell can release
multiple mediators from the same exocytic granules which work in concert to
produce a coordinated
physiological response in the target cells. These regulated exocytic cells
include neurons
(neurotransmitter release), adrenal chromaffin cells (adrenaline secretion),
pancreatic acinar cells
(digestive enzyme secretion), pancreatic (3-cells (insulin secretion), mast
cells (histamine secretion),
mammary cells (milk protein secretion), sperm (enzyme secretion), egg cells
(creation of fertilization
envelope) and adipocytes (insertion of glucose transporters into the plasma
membrane). In addition,
current theory suggests that the basic mechanisms of vesicle docking and
fusion is conserved from
yeast to the mammalian brain.
In addition, disorders involving exocytosis are known. For example,
inflammatory mediator release
from mast cells leads to a variety of disorders, including asthma. In the
United States alone, over 50
million people suffer from asthma, rhinitis, or some other form of allergy.
Therapy for allergy remains
limited to blocking the mediators released by mast cells (anti-histamines),
non-specific anti-
inflammatory agents such as steroids and mast cell stabilizers which are only
marginally effective at
limiting the symtomatology of allergy. Similarly, Chediak-Higashi Syndrome
(CHS) is a rare autosomal
recessive disease in which neutrophils, monocytes and lymphocytes and most
cells contain giant
cytoplasmic granules. Similar disorders have been described in mice, mink,
cattle, cats and killer
whales, with the murine homolog of CHS (designated beige or bg) being the best
characterized. See
Perou et at., J. Biol. Chem. 272(47):29790 (1997) and Barbosa et al., Nature
382:262 (1996).
Furthermore, it is widely believed that a wide array of psychiatric disorders
are the result of an
imbalance between neurotransmitter exocytosis and mediator reuptake.
A large number of pharmaceuticals have been designed to specifically interfere
with the exocytic
mediators primarily through blockade of their receptors. However, this
approach is limited by the fact
that a single receptor blocker cannot overcome the effects of many diverse
mediators.

Accordingly, it is an object of the present invention to provide methods for
screening for alterations in
exocytosis, particularly for screening for agents capable of mediating such
exocytosis. It is also an
object to provide such screening methods wherein assay background is reduced
and specificity is
increased.

SUMMARY OF THE INVENTION

In accordance with the objects outlined above, the present invention provides
methods for screening
3


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
bioactive agents for the ability to after or modulate alterations in cellular
phenotypes. The methods
generally comprise combining at least one candidate bioactive agent and a
population of cells, sorting
the cells in a FACS machine by separating the cells on the basis of at least
three, four or five cellular
parameters. The candidate agents can be part of a molecular library comprising
fusion nucleic acids
encoding the candidate bioactive agents.

In a further aspect, the present invention provides methods for screening for
alterations in exocytosis
of a population of cells or in single cells under different conditions or
combined with different bioactive
agents. The methods comprise sorting the cells in a FACS machine by assaying
for alterations in at
least three of the properties selected from the group consisting of light
scattering, fluorescent dye
uptake, fluorescent dye release, annexin granule binding, surface granule
enzyme activity, and the
quantity of granule specific proteins.

Also provided herein is a method for screening for a bioactive agent capable
of modulating exocytosis
in a cell. This method comprises combining a candidate bioactive agent and a
population of cells and
subjecting said cells to conditions that normally cause exocytosis. The cells
are sorted in a FACS
machine by assaying for alterations in at least three of the properties
selected from the group
consisting of light scattering, fluorescent dye uptake, fluorescent dye
release, annexin granule binding,
surface granule enzyme activity, and the quantity of granule specific
proteins. Alterations in at least
one of said properties as compared to cells that were not exposed to the
candidate bioactive agent
indicates that said agent modulates exocytosis.

In a preferred embodiment of the method for screening for a bioactive agent,
the properties selected
include at least one property selected from the group consisting of
fluorescent dye release, annexin
granule binding, surface granule enzyme activity, and the quantity of granule
specific proteins.

When fluoroscent dye uptake is detected, the dye is preferably a styryl dye.
In the case that
fluoroscent dye release is detected, the dye can be a low pH concentration dye
or a styryl dye.

In a preferred embodiment, the surface granule enzyme activity is detected by
an in situ enzymology
assay or by a population based enzyme assay. The enzyme substrate can be any
detectable
substrate. Preferably, the enzyme substrate is coupled to a FRET construct.
FRET constructs include
two fluoroscent proteins divided by a protease site. In this case, the
protease site is specific for a
granule protease.
In a preferred embodiment, granule specific proteins are detected. The granule
specific proteins can
be any detectable protein. In one embodiment, the granule specific proteins
are fusion proteins

4


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
comprising a granule specific protein and a detectable molecule which can be a
FRET construct.

In another preferred embodiment, a method for screening for a bioactive agent
capable of modulating
exocytosis in a cell is provided wherein said method comprises combining at
least one candidate
bioactive agent and a population of cells each containing a fusion nucleic
acid comprising a nucleic
acid encoding a granule-specific protein and a label. The cells are subjected
to conditions that
normally cause exocytosis and the alterations in the quantity of the label is
detected. Alterations in the
quantity of the label indicates that the agent modulates exocytosis.
Preferably, the label is an epitope
tag or a fluorescent molecule. In a preferred embodiment, the fluorescent
molecule is a FRET
construct.

In an additional aspect, the invention provides methods of screening for
exocytosis modulators
comprising combining candidate bioactive agents, cells comprising nucleic
acids encoding a
detectable granule-specific protein, and an agent for detecting this protein.
The cells are subjected to
conditions that normally cause exocytosis, and the presence or absence of the
protein is determined.
In another preferred embodiment, a method for screening for a bioactive agent
capable of modulating
exocytosis in a cell is provided which comprises combining at least one
candidate bioactive agent and
a population of cells. The cells are subjected to conditions that normally
cause exocytosis and a
fluorescent annexin is added. Alterations in the amount of the fluorescent
annexin on the surface of
the cells is evaluated.

In another preferred embodiment, a method for screening for a bioactive agent
capable of modulating
exocytosis in a cell is provided which comprises providing a population of
cells wherein the cells have
taken in a low pH concentration dye. The low pH concentration dye loaded cells
are combined with at
least one candidate bioactive agent and subjected to conditions that normally
cause exocytosis. The
release of the low pH concentration dye is detected. Alterations in the amount
of released dye indicate
that the agent modulates exocytosis.

In another preferred embodiment, a method for screening for a bioactive agent
capable of modulating
exocytosis in a cell is provided which comprises combining at least one
candidate bioactive agent and
a population of cells. The cells are subjected to conditions that normally
cause exocytosis and a
fluorescent substrate specific to a granule enzyme is added. The fluorescent
substrate specific to a
granule enzyme is detected, wherein alterations in the amount of the
fluorescent substrate indicative
that the agent modulates exocytosis. In a preferred embodiment, the substrate
comprises a FRET
construct.

5


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
In an additional aspect, the present invention provides methods and
compositions for screening for
bioactive agents capable of modulating cell cycle regulation in a cell. The
method comprises
combining a library of candidate bioactive agents and a population of cells,
sorting the cells in a FACS
machine by separating the cells on the basis of at least a cell viability
assay, a proliferation assay, and
a cell phase assay.

In a further aspect, the methods comprise expressing a library of fusion
nucleic acids in a library of
cells. The fusion nucleic acids comprise a nucleic acid encoding a candidate
bioactive agent and a
detectable moiety. The cells are sorted in a FACS machine by separating the
cells; when the cellular
phenotype is cell cycle, the cells are sorted on the basis of at least a cell
viability assay, an expression
assay, a proliferation assay, and a cell phase assay.

BRIEF DESCRIPTION OF THE DRAWINGS

Figures 1A, 113 and 1C schematically depict three retroviral constructions of
Example 1. Figure 1A
includes the CRU5-GFP-p21 construction, comprising a CRU5 promoter, the yr-
retroviral packaging
signal, the coding region for GFP, fused to the coding region of p21, followed
by an LTR. Figure 1 B
depicts the CRU5-GFP-p2lC construction, which includes the C-terminal 24 amino
acids of p21.
Figure 1 C depicts the CRU5-GFP-pUCmut construct, which is a mutant version of
CRU5-p21 C with 3
alanine substitutions.

Figures 2A, 2B, 2C and 2D depict the results of the experiments of Example 1.
Figure 2A depicts a
viability assay utilizing forward and side scatter. Cells exhibiting a
characteristic ratio are collected.
Figure 2B shows the fluorescence of the GFP of the vectors. Figure 2C depicts
the use of PKH26, an
inclusion dye, in a proliferation assay; the cells containing p21, a protein
known to arrest cells, remain
brightly fluorescent, while the control cells continue to proliferate, thus
diluting the dye and losing
fluorescence. Figure 2D depicts the use of Hoechst 33342 in a cell phase
assay.

Figure 3 depicts the effect of AraC treatment on Jurkat cells infected with
p21, an agent that arrests
cells in the GI phase. AraC is a nucleotide analog that is toxic to dividing
cells. Thus, those cells that
are cell cycle arrested survive. The lower line depicts Jurkat cells without
the p21 insert, and the upper
line depicts Jurkat cells with the p21 insert.

Figures 4A and 4B depict bar graphs showing the results of a population based
exocytic enzyme
activity assay for exocytosis. Figure 4A shows glucuronidase or hexosaminidase
activity in the
supernatant of cells combined with DMSO (-) or ionomycin (+). Figure 4B shows
hexosaminidase
activity in the supernatant of cells sensitized with varying amounts of IgE
anti-DNP and stimulated with

6


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
increasing amounts of the antigen BSA-DNP.

Figures 5A-5F show exocytic light scatter changes observed on the flow
cytometer, side scatter vs.
forward scatter, plotted as bivariate histograms for RBL-2H3 cells (Figures 5A
and 5D) and MC-9 cells
(Figures 5B, 5C, 5E and 5F). After stimulation with an ionophore, the cells
were observed at 0
minutes (Figures 5A and 5C), 5 minutes (Figure 5E), 10 minutes (Figure 5D),
and 30 minutes (Figures
5B and 5F).

Figures 6A-6E show graphs of the results of a styryl dye assay to detect
exocytosis by FAGS. Cells
were combined with (left peaks) DMSO or (right peaks) ionomycin in the
presence of either FM 4-64
(Figures 6A and 6B) or FM 1-43 (Figures 6C, 6D and 6E). Figures 6A and 6C show
cells detected in
fluorescence channel 1. Figures 6B and 6D show cells detected in fluorescence
channel 3. Figure 6E
shows the mean channel shift detected in the flow cytometer in fluorescence
channel 1 plotted as a
bar graph wherein cells were preincubated with varying doses of the PI-3
kinase inhibitor wortmannin
prior to administration of an ionophore (bars 1-4) or DMSO (bar 5) in the
presence of FM 1-43.
Figures 7A-7D show graphs depicting the results of an annexin-V detection
assay of exocytosis by
FAGS. Cells were combined with either DMSO (Figures 7A and 7B) or ionomycin
(Figures 7C and
7D) and then stained with both propidium iodide (Figures 7A and 7C) and
annexin-V-FITC (Figures 7B
and 7D).

Figures 8A-8C show graphs indicating the results of an in situ enzymology
assay of exocytosing cells
visualized by FACS. Cells were combined with DMSO (Figure 8A) or an ionophore
(Figures 8B and
8C) and then stained for in situ glucuronidase activity. Figure 8C shows the
pH profile of the cell
surface enzymatic activity wherein the bar graphs represent the percentage of
maximal signal, as
measured by mean channel shift in the flow cytometer, observed.

Figure 9 is a histogram of fluorescence intensity detected in channel 1
showing cells loaded with
LYSOTRACKER GREENTM, combined with either DMSO (left) or ionomycin (right) and
viewed in the
flow cytometer.

Figures 1 OA-1 OH show the results of a multiparameter analysis including
detection of LYSOTRACKER
GREENTM , annexin-V-APC and forward and side scatter. Figures 1OA-10D and 1OE-
10H each show
cells treated with increasing doses of ionomycin and observed in the flow
cytometer with four
parameters simultaneously. The cells were loaded with low pH concentration
dye, stimulated and
stained with annexin-V-APC. Figures 1OA-IOD show bivariate histograms of side
vs. forward light
scatter and Figures 10E-10H show bivariate histograms of annexin-V-APC vs. low
pH concentration

7


CA 02325597 2008-09-10
77245-54

dye signals.

Figure 11 shows a graph of cells stimulated in the
presence of FM 1-43 and annexin-V-APC stained. At various
timepoints after ionomycin stimulation the cells were

analyzed by flow cytometry and the supernatant for enzymatic
activity (cell supernatant). The parameters forward
scatter, FM-143, annexin-V-APC, and hexosaminidase are
plotted on the graph relative to the maximal response for
each parameter. For calcium signalling, a separate tube of

cells was loaded with Fluo-3 and underwent the identical
procedure.

DETAILED DESCRIPTION OF THE INVENTION

The present invention is directed to the detection
of alterations in cellular phenotypes, such as cell cycle

regulation, exocytosis, small molecule toxicity, cell
surface receptor expression, enzyme expression, etc. by
evaluating or assaying a variety of cellular parameters,
generally through the use of a fluorescence-activated cell
sorter (FACS) machine. There are a number of parameters

that can be measured to allow detection of alterations in a
variety of cellular phenotypes as is more fully outlined
below. By assaying a plurality of these parameters either
sequentially or preferably simultaneously, rapid and
accurate screening may be done.

In one embodiment, the invention provides a method
comprising: a) introducing a nucleic acid that encodes a
candidate bioactive agent into a population of cells to
produce contacted cells that produce said candidate

bioactive agent; and b) sorting said contacted cells on the
basis of at least five cellular parameters using a
fluorescence-activated cell sorter (FACS), and c)

8


CA 02325597 2008-09-10
77245.-54

identifying a cell having a phenotype that is altered by
said candidate bioactive agent.

In one embodiment, the invention provides a method
of screening for a bioactive agent capable of altering a

cellular phenotype, said method comprising: a) introducing a
library of nucleic acids, each nucleic acid in said library
encoding a candidate bioactive agent, into a population of
cells; and b) sorting said cells in a FACS machine by
separating said cells on the basis of at least five cellular
parameters.

In a preferred embodiment, the methods outlined
herein are used to screen for modulators of cellular
phenotypes. Cellular phenotypes that may be assayed
include, but are not limited to, cellular apoptosis,
including cell cycle regulation, exocytosis, toxicity to
small molecules, the expression of any number of moieties
including receptors (particularly cell surface receptors),
adhesion molecules, cytokine secretion, protein-protein
interactions, etc.

In a preferred embodiment, the methods are used to
evaluate cell cycle regulation. In this embodiment,
preferred cellular parameters or assays are cell viability
assays, assays to determine whether cell are arrested at a
particular cell cycle stage ("cell proliferation assays"),

and assays to determine at which cell stage the cells have
arrested ("cell phase assays"). By assaying or measuring
one or more of these parameters, it is possible to detect
not only alterations in cell cycle regulation, but

alterations of different steps of the cell cycle regulation
pathway. This may be done to evaluate native cells, for
example to quantify the aggressiveness of a tumor cell type,
or to evaluate the effect of candidate drug agents that are

8a


CA 02325597 2008-09-10
77245-54

being tested for their effect on cell cycle regulation. In
this manner, rapid, accurate screening of candidate agents
may be performed to identify agents that modulate cell cycle
regulation.

Thus, the present methods are useful to elucidate
bioactive agents that can cause a population of cells

8b


CA 02325597 2000-10-16

WO 99/54494 PCT/US"/08345
to either move out of one growth phase and into another, or arrest in a growth
phase. In some
embodiments, the cells are arrested in a particular growth phase, and it is
desirable to either get them
out of that phase or into a new phase. Alternatively, it may be desirable to
force a cell to arrest in a
phase, for example G1, rather than continue to move through the cell cycle.
Similarly, it may be
desirable in some circumstances to accelerate a non-arrested but slowly moving
population of cells
into either the next phase or just through the cell cycle, or to delay the
onset of the next phase. For
example, it may be possible to alter the activities of certain enzymes, for
example kinases,
phosphatases, proteases or ubiquitination enzymes, that contribute to
initiating cell phase changes.

In a preferred embodiment, the methods outlined herein are done on cells that
are not arrested in the
G1 phase; that is, they are rapidly or uncontrollably growing and replicating,
such as tumor cells. In
this manner, candidate agents are evaluated to find agents that can alter the
cell cycle regulation, i.e.
cause the cells to arrest at cell cycle checkpoints, such as in G1 (although
arresting in other phases
such as S, G2 or M are also desirable). Alternatively, candidate agents are
evaluated to find agents
that can cause proliferation of a population of cells, i.e. that allow cells
that are generally arrested in
G1 to start proliferating again; for example, peripheral blood cells,
terminally differentiated cells, stem
cells in culture, etc.

Accordingly, in a preferred embodiment, the invention provides methods for
screening for alterations in
cell cycle regulation of a population of cells. "Alteration" and "modulation"
(used herein
interchangeably), as used herein can include both increases and decreases in
the parameter or
phenotype being measured. By "alteration" or "modulation" in the context of
cell cycle regulation, is
generally meant one of two things. In a preferred embodiment, the alteration
results in a change in the
cell cycle of a cell, i.e. a proliferating cell arrests in any one of the
phases, or an arrested cell moves
out of its arrested phase and starts the cell cycle, as compared to another
cell or in the same cell
under different conditions. Alternatively, the progress of a cell through any
particular phase may be
altered; that is, there may be an acceleration or delay in the length of time
it takes for the cells to move
thorough a particular growth phase. For example, the cell may be normally
undergo a G1 phase of
several hours; the addition of an agent may prolong the G1 phase.
The measurements can be determined wherein all of the conditions are the same
for each
measurement, or under various conditions, with or without bioactive agents, or
at different stages of
the cell cycle process. For example, a measurement of cell cycle regulation
can be determined in a
cell population wherein a candidate bioactive agent is present and wherein the
candidate bioactive
agent is absent. In another example, the measurements of cell cycle regulation
are determined
wherein the condition or environment of the populations of cells differ from
one another. For example,
the cells may be evaluated in the presence or absence of physiological
signals, for example

9


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
hormones, antibodies, peptides, antigens, cytokines, growth factors, action
potentials,
pharmacological agents (i.e. chemotherapeutics, etc.), or other cells (i.e.
cell-cell contacts). In another
example, the measurements of cell cycle regulation are determined at different
stages of the cell cycle
process. In yet another example, the measurements of cell cycle regulation are
taken wherein the
conditions are the same, and the alterations are between one cell or cell
population and another cell or
cell population.

By a "population of cells" or "library of cells" or "plurality of cells"
herein is meant at least two cells, with
at least about 103 being preferred, at least about 106 being particularly
preferred, and at least about
108 to 109 being especially preferred. The population or sample can contain a
mixture of different cell
types from either primary or secondary cultures although samples containing
only a single cell type are
preferred, for example, the sample can be from a cell line, particularly tumor
cell lines (particularly
when , as outlined below. The cells may be in any cell phase, either
synchronously or not, including
M, G1, S, and G2. In a preferred embodiment, cells that are replicating or
proliferating are used; this
may allow the use of retroviral vectors for the introduction of candidate
bioactive agents. Alternatively,
non-replicating cells may be used, and other vectors (such as adenovirus and
lentivirus vectors) can
be used. In addition, although not required, the cells are compatible with
dyes and antibodies.
Preferred cell types for use in the invention will vary with the cellular
phenotype to be modulated.
Suitable cells include, but are not limited to, mammalian cells, including
animal (rodents, including
mice, rats, hamsters and gerbils), primates, and human cells, particularly
including tumor cells of all
types, including breast, skin, lung, cervix, colonrectal, leukemia, brain,
etc. As outlined below,
additional cell types may be used for screening for exocytosis.

In a preferred embodiment, the cell cycle regulation methods comprise sorting
the cells in a FACS
machine by assaying several different cell parameters, including, but not
limited to, cell viability, cell
proliferation, and cell phase.

In a preferred embodiment, cell viability is assayed, to ensure that a lack of
cellular change is due to
experimental conditions (i.e. the introduction of a candidate bioactive agent)
not cell death. There are
a variety of suitable cell viability assays which can be used, including, but
not limited to, light
scattering, viability dye staining, and exclusion dye staining.

In a preferred embodiment, a light scattering assay is used as the viability
assay, as is well known in
the art. When viewed in the FACS, cells have particular characteristics as
measured by their forward
and 90 degree (side) light scatter properties. These scatter properties
represent the size, shape and
granule content of the cells. These properties account for two parameters to
be measured as a



CA 02325597 2008-09-10
,77245-54

readout for the viability. Briefly, the DNA of dying or dead cells generally
condenses, which alters the
90 scatter; similarly, membrane blebbing can alter the forward scatter.
Alterations in the intensity of
light scattering, or the cell-refractive index indicate alterations in
viability.

Thus, in general, for light scattering assays, a live cell population of a
particular cell type is evaluated
to determine it's forward and side scattering properties. This sets a standard
for scattering that can
subsequently be used.

In a preferred embodiment, the viability assay utilizes a viability dye. There
are a number of known
viability dyes that stain dead or dying cells, but do not stain growing cells.
For example, annexin V is a
member of a protein family which displays specific binding to phospholipid
(phosphotidylserine) in a
divalent ion dependent manner. This protein has been widely used for the
measurement of apoptosis
(programmed cell death) as cell surface exposure of phosphatidylserine is a
hallmark early signal of
this process. Suitable viability dyes include, but are not limited to,
annexin, ethidium homodimer-1,
DEAD Red, propidium iodide, SYTOX Green, etc., and others known in the art;
see the Molecular
Probes Handbook of Fluorescent Probes and Research Chemicals, Haugland, Sixth
Edition;
see Apoptosis Assay on page 285 in particular, and Chapter 16.

Protocols for viability dye staining for cell viability are known, see
Molecular Probes catalog, supra. In
this embodiment, the viability dye such as annexin is labeled, either directly
or indirectly, and
combined with a cell population. Annexin is commercially available, i.e., from
PharMingen, San Diego,
California, or Caltag Laboratories, Millbrae, California. Preferably, the
viability dye is provided in a
solution wherein the dye is in a concentration of about 100 ng/ml to about 500
ng/ml, more preferably,
about 500 ng/ml to about 1 pg/ml, and most preferably, from about 1 pg/ml to
about 5 pg/ml. In a
preferred embodiment; the viability dye is directly labeled; for example,
annexin may be labeled with a
fluorochrome such as fluorecein isothiocyanate (FITC), Alexa dyes, TRITC,
AMCA, APC, tri-color, Cy-
5, and others known in the art or commercially available. In an alternate
preferred embodiment, the
viability dye is labeled with a first label, such as a hapten such as biotin,
and a secondary fluorescent
label is used, such as fluorescent streptavidin. Other first and second
labeling pairs can be used as
will be appreciated by those in the art.

Once added, the viability dye is allowed to incubate with the cells for a
period of time, and washed, if
necessary. The cells are then sorted as outlined below to remove the non-
viable cells.

In a preferred embodiment, exclusion dye staining is used as the viability
assay. Exclusion dyes are
those which are excluded from living cells, i.e. they are not taken up
passively (they do not permeate
the cell membrane of a live cell). However, due to the permeability of dead or
dying cells, they are

11


CA 02325597 2008-09-10
77245-54

taken up by dead cells. Generally, but not always, the exclusion dyes bind to
DNA, for example via
intercalation. Preferably, the exclusion dye does not fluoresce, or fluoresces
poorly, in the absence of
DNA; this eliminates the need for a wash step. Alternatively, exclusion dyes
that require the use of a
secondary label may also be used. Preferred exclusion dyes include, but are
not limited to, ethidium
bromide; ethidium homodimer-1; propidium iodine; SYTOX green nucleic acid
stain; Calcein AM,
BCECF AM; fluorescein diacetate; TOTO and TO-PROTM (from Molecular Probes;
supra, see
chapter 16) and others known in the art.

Protocols for exclusion dye staining for cell viability are known, see the
Molecular Probes catalog,
supra. In general, the exclusion dye is added to the cells at a concentration
of from about 100 ng/mI to
about 500 ng/ml, more preferably, about 500 ng/ml to about 1 pg/ml, and most
preferably, from about
0.1 pg/mI to about 5 pg/ml, with about 0.5 pg/ml being particularly preferred.
The cells and the
exclusion dye are incubated for some period of time, washed, if necessary, and
then the cells sorted
as outlined below, to remove non-viable-cells from the population.
In addition, there are other cell viability assays which may be run, including
for example enzymatic
assays, which can measure extracellular enzymatic activity of either live
cells (i.e. secreted proteases,
etc.), or dead cells (i.e. the presence of intracellular enzymes in the media;
for example, intracellular
proteases, mitochondrial enzymes, etc.). See the Molecular Probes Handbook of
Fluorescent Probes
and Research Chemicals, Haugland, Sixth Edition, see chapter 16
in particular.

In a preferred embodiment, at least one cell viability assay is run, with at
least two different cell viability
assays being preferred, when the fluors are compatible. When only 1 viability
assay is run, a preferred
embodiment utilizes light scattering assays (both forward and side
scattering). When two viability
assays are run, preferred embodiments utilize light scattering and dye
exclusion, with light scattering
and viability dye staining also possible, and all three being done in some
cases as well. Viability
assays thus allow the separation of viable cells from non-viable or dying
cells.

In addition to a cell viability assay, a preferred embodiment utilizes a cell
proliferation assay. By
"proliferation assay" herein is meant an assay that allows the determination
that a cell population is
either proliferating, i.e. replicating, or not replicating.

In a preferred embodiment, the proliferation assay is a dye inclusion assay. A
dye inclusion assay
relies on dilution effects to distinguish between cell phases. Briefly, a dye
(generally a fluorescent dye
as outlined below) is introduced to cells and taken up by the cells. Once
taken up, the dye is trapped in
the cell, and does not diffuse out. As the cell population divides, the dye is
proportionally diluted. That
12


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
is, after the introduction of the inclusion dye, the cells are allowed to
incubate for some period of time;
cells that lose fluorescence over time are dividing, and the cells that remain
fluorescent are arrested in
a non-growth phase.

Generally, the introduction of the inclusion dye may be done in one of two
ways. Either the dye cannot
passively enter the cells (e.g. it is charged), and the cells must be treated
to take up the dye; for
example through the use of a electric pulse. Alternatively, the dye can
passively enter the cells, but
once taken up, it is modified such that it cannot diffuse out of the cells.
For example, enzymatic
modification of the inclusion dye may render it charged, and thus unable to
diffuse out of the cells. For
example, the Molecular Probes CellTrackerTm dyes are fluorescent chloromethyl
derivatives that freely
diffuse into cells, and then glutathione S-transferase-mediated reaction
produces membrane
impermeant dyes.

Suitable inclusion dyes include, but are not limited to, the Molecular Probes
line of CellTrackerTm dyes
, including, but not limited to CellTrackerTm Blue, CellTrackerTM Yellow-
Green, CellTrackerTM Green,
CellTrackerTM Orange, PKH26 (Sigma), and others known in the art; see the
Molecular Probes
Handbook, supra; chapter 15 in particular.

In general, inclusion dyes are provided to the cells at a concentration
ranging from about
100 ng/ml to about 5 pg/mi, with from about 500 ng/ml to about 1 pg/ml being
preferred. A wash step
may or may not be used. In a preferred embodiment, a candidate bioactive agent
is combined with the
cells as described herein. The cells and the inclusion dye are incubated for
some period of time, to
allow cell division and thus dye dilution. The length of time will depend on
the cell cycle time for the
particular cells; in general, at least about 2 cell divisions are preferred,
with at least about 3 being
particularly preferred and at least about 4 being especially preferred. The
cells are then sorted as
outlined below, to create populations of cells that are replicating and those
that are not. As will be
appreciated by those in the art, in some cases, for example when screening for
anti-proliferation
agents, the bright (i.e. fluorescent) cells are collected; in other
embodiments, for example for
screening for proliferation agents, the low fluorescence cells are collected.
Alterations are determined
by measuring the fluorescence at either different time points or in different
cell populations, and
comparing the determinations to one another or to standards.

In a preferred embodiment, the proliferation assay is an antimetabolite assay.
In general,
antimetabolite assays find the most use when agents that cause cellular arrest
in G1 or G2 resting
phase is desired. In an antimetabolite proliferation assay, the use of a toxic
antimetabolite that will kill
dividing cells will result in survival of only those cells that are not
dividing. Suitable antimetabolites
include, but are not limited to, standard chemotherapeutic agents such as
methotrexate, cisplatin,

13


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
taxol, hydroxyurea, nucleotide analogs such as AraC, etc. In addition,
antimetabolite assays may
include the use of genes that cause cell death upon expression.

The concentration at which the antimetabolite is added will depend on the
toxicity of the particular
antimetabolite, and will be determined as is known in the art. The
antimetabolite is added and the cells
are generally incubated for some period of time; again, the exact period of
time will depend on the
characteristics and identity of the antimetabolite as well as the cell cycle
time of the particular cell
population. Generally, a time sufficient for at least one cell division to
occur.

In a preferred embodiment, at least one proliferation assay is run, with more
than one being preferred.
Thus, a proliferation assay results in a population of proliferating cells and
a population of arrested
cells.

In a preferred embodiment, either after or simultaneously with one or more of
the proliferation assays
outlined above, at least one cell phase assay is done. A "cell phase" assay
determines at which cell
phase the cells are arrested, M, GI, S, or G2.

In a preferred embodiment, the cell phase assay is a DNA binding dye assay.
Briefly, a DNA binding
dye is introduced to the cells, and taken up passively. Once inside the cell,
the DNA binding dye binds
to DNA, generally by intercalation, although in some cases, the dyes can be
either major or minor
groove binding compounds. The amount of dye is thus directly correlated to the
amount of DNA in the
cell, which varies by cell phase; G2 and M phase cells have twice the DNA
content of G1 phase cells,
and S phase cells have an intermediate amount, depending on at what point in S
phase the cells are.
Suitable DNA binding dyes are permeant, and include, but are not limited to,
Hoechst 33342 and
33258, acridine orange, 7-AAD, LDS 751, DAPI, and SYTO 16, Molecular Probes
Handbook, supra;
chapters 8 and 16 in particular.

In general, the DNA binding dyes are added in concentrations ranging from
about 1 pg/ml to about 5
pg/ml. The dyes are added to the cells and allowed to incubate for some period
of time; the length of
time will depend in part on the dye chosen. In one embodiment, measurements
are taken immediately
after addition of the dye. The cells are then sorted as outlined below, to
create populations of cells
that contain different amounts of dye, and thus different amounts of DNA; in
this way, cells that are
replicating are separated from those that are not. As will be appreciated by
those in the art, in some
cases, for example when screening for anti-proliferation agents, cells with
the least fluorescence (and
thus a single copy of the genome) can be separated from those that are
replicating and thus contain
more than a single genome of DNA. Alterations are determined by measuring the
fluorescence at
either different time points or in different cell populations, and comparing
the determinations to one
14


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
another or to standards.

In a preferred embodiment, the cell phase assay is a cyclin destruction assay.
In this embodiment,
prior to screening (and generally prior to the introduction of a candidate
bioactive agent, as outlined
below), a fusion nucleic acid is introduced to the cells. The fusion nucleic
acid comprises nucleic acid
encoding a cyclin destruction box and a nucleic acid encoding a detectable
molecule. "Cyclin
destruction boxes" are known in the art and are sequences that cause
destruction via the
ubiquitination pathway of proteins containing the boxes during particular cell
phases. That is, for
example, G1 cyclins may be stable during G1 phase but degraded during S phase
due to the
presence of a G1 cyclin destruction box. Thus, by linking a cyclin destruction
box to a detectable
molecule, for example green fluorescent protein, the presence or absence of
the detectable molecule
can serve to identify the cell phase of the cell population. In a preferred
embodiment, multiple boxes
are used, preferably each with a different fluor, such that detection of the
cell phase can occur.

A number of cyclin destruction boxes are known in the art, for example, cyclin
A has a destruction box
comprising the sequence RTVLGVIGD; the destruction box of cyclin B1 comprises
the sequence
RTALGDIGN. See Glotzer et al., Nature 349:132-138 (1991). Other destruction
boxes are known as
well: YMTVSIIDRFMQDSCVPKKMLQLVGVT (rat cyclin B); KFRLLQETMYMTVSIIDRFMQNSCVPKK
(mouse cyclin B); RAILIDWLIQVQMKFRLLQETMYMTVS (mouse cyclin B1);
DRFLQAQLVCRKKLQWGITALLLASK (mouse cyclin B2); and MSVLRGKLQLVGTAAMLL (mouse
cyclin A2).

The nucleic acid encoding the cyclin destruction box is operably linked to
nucleic acid encoding a
detectable molecule. The fusion proteins are constructed by methods known in
the art. For example,
the nucleic acids encoding the destruction box is ligated to a nucleic acid
encoding a detectable
molecule. By "detectable molecule" herein is meant a molecule that allows a
cell or compound
comprising the detectable molecule to be distinguished from one that does not
contain it, i.e., an
epitope, sometimes called an antigen TAG, a specific enzyme, or a fluorescent
molecule. Preferred
fluorescent molecules include but are not limited to green fluorescent protein
(GFP), blue fluorescent
protein (BFP), yellow fluorescent protein (YFP), red fluorescent protein
(RFP), and enzymes including
luciferase and a-galactosidase. When antigen TAGs are used, preferred
embodiments utilize cell
surface antigens. The epitope is preferably any detectable peptide which is
not generally found on the
cytoplasmic membrane, although in some instances, if the epitope is one
normally found on the cells,
increases may be detected, although this is generally not preferred.
Similarly, enzymatic detectable
molecules may also be used; for example, an enzyme that generates a novel or
chromogenic product.
Accordingly, the results of sorting after cell phase assays generally result
in at least two populations of


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
cells that are in different cell phases.

In a preferred embodiment, the methods are used to screen candidate bioactive
agents for the ability
to modulate cell cycle regulation, including the activation or suppression of
cell cycle checkpoint
pathways and ameliorating checkpoint defects. The candidate bioactive agent
can be added to the
cell population exogenously or can be introduced into the cells as described
further herein.

The term "candidate bioactive agent" or "exogeneous compound" as used herein
describes any
molecule, e.g., protein, small organic molecule, carbohydrates (including
polysaccharides),
polynucleotide, lipids, etc. Generally a plurality of assay mixtures are run
in parallel with different
agent concentrations to obtain a differential response to the various
concentrations. Typically, one of
these concentrations serves as a negative control, i.e., at zero concentration
or below the level of
detection. In addition, positive controls can be used. For example, in the
cell cycling assays, agents
known to alter cell cycling may be used. For example, p21 is a molecule known
to arrest cells in the
G1 cell phase, by binding G1 cyclin-CDK complexes. Similarly, for exocytosis,
compounds known to
induce exocytosis can be used as is more fully outlined below.

Candidate agents encompass numerous chemical classes, though typically they
are organic
molecules, preferably small organic compounds having a molecular weight of
more than 100 and less
than about 2,500 daltons. Candidate agents comprise functional groups
necessary for structural
interaction with proteins, particularly hydrogen bonding, and typically
include at least an amine,
carbonyl, hydroxyl or carboxyl group, preferably at least two of the
functional chemical groups. The
candidate agents often comprise cyclical carbon or heterocyclic structures
and/or aromatic or
polyaromatic structures substituted with one or more of the above functional
groups. Candidate
agents are also found among biomolecules including peptides, saccharides,
fatty acids, steroids,
purines, pyrimidines, derivatives, structural analogs or combinations thereof.
Particularly preferred are
peptides.

Candidate agents are obtained from a wide variety of sources including
libraries of synthetic or natural
compounds. For example, numerous means are available for random and directed
synthesis of a
wide variety of organic compounds and biomolecules, including expression of
randomized
oligonucleotides. Alternatively, libraries of natural compounds in the form of
bacterial, fungal, plant
and animal extracts are available or readily produced. Additionally, natural
or synthetically produced
libraries and compounds are readily modified through conventional chemical,
physical and biochemical
means. Known pharmacological agents may be subjected to directed or random
chemical
modifications, such as acylation, alkylation, esterification, amidification to
produce structural analogs.
16


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
In a preferred embodiment, the candidate bioactive agents are proteins. By
"protein" herein is meant
at least two covalently attached amino acids, which includes proteins,
polypeptides, oligopeptides and
peptides. The protein may be made up of naturally occurring amino acids and
peptide bonds, or
synthetic peptidomimetic structures. Thus "amino acid", or "peptide residue",
as used herein means
both naturally occurring and synthetic amino acids. For example, homo-
phenylalanine, citrulline and
noreleucine are considered amino acids for the purposes of the invention.
"Amino acid" also includes
imino acid residues such as proline and hydroxyproline. The side chains may be
in either the (R) or
the (S) configuration. In the preferred embodiment, the amino acids are in the
(S) or L-configuration.
If non-naturally occurring side chains are used, non-amino acid substituents
may be used, for example
to prevent or retard in vivo degradations. Chemical blocking groups or other
chemical substituents
may also be added.

In a preferred embodiment, the candidate bioactive agents are naturally
occurring proteins or
fragments of naturally occurring proteins. Thus, for example, cellular
extracts containing proteins, or
random or directed digests of proteinaceous cellular extracts, may be used. In
this way libraries of
procaryotic and eukaryotic proteins may be made for screening in the systems
described herein.
Particularly preferred in this embodiment are libraries of bacterial, fungal,
viral, and mammalian
proteins, with the latter being preferred, and human proteins being especially
preferred.

In a preferred embodiment, the candidate bioactive agents are peptides of from
about 5 to about 30
amino acids, with from about 5 to about 20 amino acids being preferred, and
from about 7 to about 15
being particularly preferred. The peptides may be digests of naturally
occurring proteins as is outlined
above, random peptides, or "biased" random peptides. By "randomized" or
grammatical equivalents
herein is meant that each nucleic acid and peptide consists of essentially
random nucleotides and
amino acids, respectively. Since generally these random peptides (or nucleic
acids, discussed below)
are chemically synthesized, they may incorporate any nucleotide or amino acid
at any position. The
synthetic process can be designed to generate randomized proteins or nucleic
acids, to allow the
formation of all or most of the possible combinations over the length of the
sequence, thus forming a
library of randomized candidate bioactive proteinaceous agents.
In one embodiment, the library is fully randomized, with no sequence
preferences or constants at any
position. In a preferred embodiment, the library is biased. That is, some
positions within the sequence
are either held constant, or are selected from a limited number of
possibilities. For example, in a
preferred embodiment, the nucleotides or amino acid residues are randomized
within a defined class,
for example, of hydrophobic amino acids, hydrophilic residues, sterically
biased (either small or large)
residues, towards the creation of cysteines, for cross-linking, prolines for
SH-3 domains, serines,
threonines, tyrosines or histidines for phosphorylation sites, etc., or to
purines, etc.

17


CA 02325597 2008-09-10
77245-54

In a preferred embodiment, the candidate bioactive agents are nucleic acids.
By "nucleic acid" or
", oligonucleotide" or grammatical equivalents herein means at least two
nucleotides covalently linked
together. A nucleic acid of the present invention will generally contain
phosphodiester bonds, although
in some cases, as outlined below, nucleic acid analogs are included that may
have alternate
backbones, comprising, for example, phosphoramide (Beaucage, et al.,
Tetrahedron, 49(10):1925
(1993) and references therein; Letsinger, J. Org. Chem., 35:3800 (1970);
Sprinzl, et al., Eur. J.
Biochem., 81:579 (1977); Letsinger, et aL, Nucl. Acids Res., 14:3487 (1986);
Sawai, eta!., Chem.
Lett.; 805 (1984), Letsinger, et al., J. Am. Chem. Soc., 110:4470 (1988); and
Pauwels, et al., Chemica
Scri ta, 26:141 (1986)), phosphorothioate (Mag, et al., Nucleic Acids Res.,
19:1437 (1991); and U.S.
Patent No. 5,644,048), phosphorodithioate (Briu, et aL, J. Am. Chem. Soc.,
111:2321 (1989)), O-
methylphophoroamidite linkages (see Eckstein, Oligonucleotides and Analogues:
A Practical
Approach, Oxford University Press), and peptide nucleic acid backbones and
linkages (see Egholm, J.
Am. Chem. Soc., 114:1895 (1992); Meier, et aL, Chem. Int. Ed. Engl., 31:1008
(1992); Nielsen,
Nature, 365:566 (1993); Carlsson, et al., Nature, 380:207 (1996).
Other analog nucleic acids include those with positive backbones (Denpcy, et
al., Proc.
Natl. Acad. Sci. USA, 92:6097 (1995)); non-ionic backbones (U.S. Patent Nos.
5,386,023; 5,637,684;
5,602,240; 5,216,141; and 4,469,863; Kiedrowshi, et al., Angew. Chem. Intl.
Ed. English, 30:423
(1991); Letsinger, et al., J. Am. Chem.. Soc., 110:4470 (1988); Letsinger, et
al., Nucleoside &
Nucleotide, 13:1597 (1994); Chapters 2 and 3, ASC Symposium Series 580,
"Carbohydrate
Modifications in Antisense Research", Ed. Y.S. Sanghui and P. Dan Cook;
Mesmaeker, et al.,
Bioorganic & Medicinal Chem. Left., 4:395 (1994); Jeffs, et al., J.
Biomolecular NMR, 34:17 (1994);
Tetrahedron Lett., 37:743 (1996)) and non-ribose backbones, including those
described in U.S. Patent
Nos. 5,235,033 and 5,034,506, and Chapters 6 and 7, ASC Symposium Series 580,
"Carbohydrate
Modifications in Antisense Research", Ed. Y.S. Sanghui and P. Dan Cook.
Nucleic acids containing
one or more carbocyclic sugars are also included within the definition of
nucleic acids (see Jenkins, et
a!, Chem. Soc. Rev., (1995) pp. 169-176). Several nucleic acid analogs are
described in Rawls, C &
E News, June 2, 1997, page 35.
These modifications of the ribose-phosphate backbone may be done to facilitate
the
addition of additional moieties such as labels, or to increase the stability
and half-life of such molecules
in physiological environments. In addition, mixtures of naturally occurring
nucleic acids and analogs
can be made. Alternatively, mixtures of different nucleic acid analogs, and
mixtures of naturally
occurring nucleic acids and analogs may be made. The nucleic acids may be
single stranded or
double stranded, as specified, or contain portions of both double stranded or
single stranded
sequence. The nucleic acid may be DNA, both genomic and cDNA, RNA or a hybrid,
where the
nucleic acid contains any combination of deoxyribo- and ribo-nucleotides, and
any combination of
bases, including uracil, adenine, thymine, cytosine, guanine, inosine,
xathanine hypoxathanine,
isocytosine, isoguanine, etc.

18


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
As described above generally for proteins, nucleic acid candidate bioactive
agents may be naturally
occurring nucleic acids, random nucleic acids, or "biased" random nucleic
acids. For example, digests
of procaryotic or eukaryotic genomes may be used as is outlined above for
proteins.

In a preferred embodiment, the candidate bioactive agents are organic chemical
moieties, a wide
variety of which are available in the literature.

In a preferred embodiment, a library of different candidate bioactive agents
are used. Preferably, the
library should provide a sufficiently structurally diverse population of
randomized agents to effect a
probabilistically sufficient range of diversity to allow binding to a
particular target. Accordingly, an
interaction library should be large enough so that at least one of its members
will have a structure that
gives it affinity for the target. Although it is difficult to gauge the
required absolute size of an inter-
action library, nature provides a hint with the immune response: a diversity
of 10'-108 different antibod-
ies provides at least one combination with sufficient affinity to interact
with most potential antigens
faced by an organism. Published in vitro selection techniques have also shown
that a library size of
107 to 108 is sufficient to find structures with affinity for the target. A
library of all combinations of a
peptide 7 to 20 amino acids in length, such as generally proposed herein, has
the potential to code for
20' (109) to 2020. Thus, with libraries of 107 to 108 different molecules the
present methods allow a
"working" subset of a theoretically complete interaction library for 7 amino
acids, and a subset of
shapes for the 2020 library. Thus, in a preferred embodiment, at least 106,
preferably at least 107,
more preferably at least 108 and most preferably at least 109 different
sequences are simultaneously
analyzed in the subject methods. Preferred methods maximize library size and
diversity.

The candidate bioactive agents are combined or added to a cell or population
of cells. Suitable cell
types for different embodiments are outlined above. The candidate bioactive
agent and the cells are
combined. As will be appreciated by those in the art, this may accomplished in
any number of ways,
including adding the candidate agents to the surface of the cells, to the
media containing the cells, or
to a surface on which the cells are growing or in contact with; adding the
agents into the cells, for
example by using vectors that will introduce the agents into the cells (i.e.
when the agents are nucleic
acids or proteins).

In a preferred embodiment, the candidate bioactive agents are either nucleic
acids or proteins
(proteins in this context includes proteins, oligopeptides, and peptides) that
are introduced into the
host cells using vectors, including viral vectors. The choice of the vector,
preferably a viral vector, will
depend on the cell type. When the cells are replicating, retroviral vectors
are used as is more fully
described below. When the cells are not replicating (i.e. they are arrested in
one of the growth
phases), other viral vectors may be used, including lentiviral and adenoviral
vectors.

19


CA 02325597 2008-09-10
77245-54

In a preferred embodiment, the cells are either replicating or can be induced
to replicate, and retroviral
vectors are used to introduce candidate bioactive agents to the cells, as is
generally outlined in PCT
US97/01019 and PCT US97/01048. Generally,
a library of retroviral vectors is made using retroviral packaging cell lines
that are helper-defective and
are capable of producing all the necessary trans proteins, including gag, pol
and env, and RNA
molecules that have in cis the q packaging signal. Briefly, the library is
generated.in a retrovirus DNA
construct backbone; standard oligonucleotide synthesis is done to generate
either the candidate agent
or nucleic acid encoding a protein, for example a random peptide, using
techniques well known in the
art. After generation of the DNA library, the library is cloned into a first
primer. The first primer serves
as a "cassette", which is inserted into the retroviral construct. The first
primer generally contains a
number of elements, including for example, the required regulatory sequences
(e.g. translation,
transcription, promoters, etc), fusion partners, restriction endonuclease
(cloning and subcloning) sites,
stop codons (preferably in all three frames), regions of complementarity for
second strand priming
(preferably at the end of the stop codon region as minor deletions or
insertions may occur in the
15. random region), etc.

A second primer is then added, which generally consists of some or all of the
complementarity region
to prime the first primer and optional necessary sequences for a second unique
restriction site for
subcloning. DNA polymerase is added to make double-stranded oligonucleotides.
The double-
stranded oligonucleotides are cleaved with the appropriate subcloning
restriction endonucleases and
subcloned into the target retroviral vectors, described below.

Any number of suitable retroviral vectors may be used. Generally, the
retroviral vectors may include:
selectable marker genes as is more fully described below; promoters driving
expression of a second
gene, placed in sense or anti-sense relative to the 5' LTR; CRU5 (a synthetic
LTR), tetracycline
regulation elements in SIN, cell specific promoters, etc.

Preferred retroviral vectors include a vector based on the murine stem cell
virus (MSCV) (see Hawley
et at., Gene Therapy 1:136 (1994)) and a modified MFG virus (Rivere et al.,
Genetics 92:6733 (1995)),
and pBABE, outlined in PCT US97/01019.

The retroviruses may include inducible and constitutive promoters for the
expression of the candidate
agent. For example, there are situations wherein it is necessary to induce
peptide expression only
during certain phases of the selection process, or only in certain cell phases
(i.e. using phase specific
promoters, such as E2F responsive promoter, p53 responsive promoter, cyclin
promoters, etc.). A
large number of both inducible and constitutive promoters are known.



CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
In addition, it is possible to configure a retroviral vector to allow
inducible expression of retroviral
inserts after integration of a single vector in target cells; importantly, the
entire system is contained
within the single retrovirus. Tet-inducible retroviruses have been designed
incorporating the Self
Inactivating (SIN) feature of 3' LTR enhancer/promoter retroviral deletion
mutant (Hoffman et al.,
PNAS USA 93:5185 (1996)). Expression of this vector in cells is virtually
undetectable in the presence
of tetracycline or other active analogs. However, in the absence of Tet,
expression is turned on to
maximum within 48 hours after induction, with uniform increased expression of
the whole population of
cells that harbor the inducible retrovirus, indicating that expression is
regulated uniformly within the
infected cell population. A similar, related system uses a mutated Tet DNA-
binding domain such that it
bound DNA in the presence of Tet, and was removed in the absence of Tet.
Either of these systems
is suitable.

In a preferred embodiment, the candidate bioactive agents are linked to a
fusion partner. By "fusion
partner" or "functional group" herein is meant a sequence that is associated
with the candidate
bioactive agent, that confers upon all members of the library in that class a
common function or ability.
Fusion partners can be heterologous (i.e. not native to the host cell), or
synthetic (not native to any
cell). Suitable fusion partners include, but are not limited to: a)
presentation structures, as defined
below, which provide the candidate bioactive agents in a conformationally
restricted or stable form; b)
targeting sequences, defined below, which allow the localization of the
candidate bioactive agent into a
subcellular or extracellular compartment; c) rescue sequences as defined
below, which allow the
purification or isolation of either the candidate bioactive agents or the
nucleic acids encoding them; d)
stability sequences, which confer stability or protection from degradation to
the candidate bioactive
agent or the nucleic acid encoding it, for example resistance to proteolytic
degradation; e) dimerization
sequences, to allow for peptide dimerization; or f) any combination of a), b),
c), d), and e), as well as
linker sequences as needed.

In a preferred embodiment, the fusion partner is a presentation structure. By
"presentation structure"
or grammatical equivalents herein is meant a sequence, which, when fused to
candidate bioactive
agents, causes the candidate agents to assume a conformationally restricted
form. Proteins interact
with each other largely through conformationally constrained domains. Although
small peptides with
freely rotating amino and carboxyl termini can have potent functions as is
known in the art, the
conversion of such peptide structures into pharmacologic agents is difficult
due to the inability to
predict side-chain positions for peptidomimetic synthesis. Therefore the
presentation of peptides in
conformationally constrained structures will benefit both the later generation
of pharmaceuticals and
will also likely lead to higher affinity interactions of the peptide with the
target protein. This fact has
been recognized in the combinatorial library generation systems using
biologically generated short
peptides in bacterial phage systems. A number of workers have constructed
small domain molecules

21


CA 02325597 2008-09-10
7724-54

in which one might present randomized peptide structures.

While the candidate bioactive agents may be either nucleic acid or peptides,
presentation structures
are preferably used with peptide candidate agents. Thus, synthetic
presentation structures, i.e.
artificial polypeptides, are capable of presenting a randomized peptide as a
conformationally-restricted
domain. Generally such presentation structures comprise a first portion joined
to the N-terminal end of
the randomized peptide, and a second portion joined to the C-terminal end of
the peptide; that is, the
peptide is inserted into the presentation structure, although variations may
be made, as outlined
below. To increase the functional isolation of the randomized expression
product, the presentation
structures are selected or designed to have minimal biologically activity when
expressed in the target
cell.

Preferred presentation structures maximize accessibility to the peptide by
presenting it on an exterior
loop. Accordingly, suitable presentation structures include, but are not
limited to, minibody structures,
loops on beta-sheet turns and coiled-coil stem structures in which residues
not critical to structure are
randomized, zinc-finger domains, cysteine-linked (disulfide) structures,
transglutaminase linked
structures, cyclic peptides, B-loop structures, helical barrels or bundles,
leucine zipper motifs, etc.
In a preferred embodiment, the presentation structure is a coiled-coil
structure, allowing the
presentation of the randomized peptide on an exterior loop. See, for example,
Myszka et al.,
Biochem. 33:2362-2373 (1994). Using this system investigators
have isolated peptides capable of high affinity interaction with the
appropriate target. In general,
coiled-coil structures allow for between 6 to 20 randomized positions.

A preferred coiled-coil presentation structure is as follows:
MGCAALESEVSALESEVASLESEVAALGRGDMPLAAVKSKLSAVKSKLASVKSKLAACGPP. The
underlined regions represent a coiled-coil leucine zipper region defined
previously (see Martin et at.,
EMBO J. 13(22):5303-5309 (1994)). The bolded GRGDMP region
represents the loop structure and when appropriately replaced with randomized
peptides (i.e.
candidate bioactive agents, generally depicted herein as (X),,, where X is an
amino acid residue and n
is an integer of at least 5 or 6) can be of variable length. The replacement
of the bolded region is
facilitated by encoding restriction endonuclease sites in the underlined
regions, which allows the direct
incorporation of randomized oligonucleotides at these positions. For example,
a preferred
embodiment generates a Xhol site at the double underlined LE site and a
Hindlll site at the double-
underlined KL site.

In a preferred embodiment, the presentation structure is a minibody structure.
A "minibody" is
22


CA 02325597 2008-09-10
77245-54

essentially composed of a minimal antibody complementarity region. The
minibody presentation
structure generally provides two randomizing-regions that in the folded
protein are presented along a
single face of the tertiary structure. See for example Bianchi et al., J. Mol.
Biol. 236(2):649-59 (1994),
and references cited therein. Investigators have shown
this minimal domain is stable in solution and have used phage selection
systems in combinatorial
libraries to select minibodies with peptide regions exhibiting high affinity,
Kd = 10', for the pro-
inflammatory cytokine IL-6.

A preferred minibody presentation structure is as follows:
MGRNSQATSGFTFSHFYMEWVRGGEYIAASRHKHNKYTTEYSASVKGRYIVSRDTSQSILYLQKKKG
PP. The bold, underline regions are the regions which may be randomized. The
italized phenylalanine
must be invariant in the first randomizing region. The entire peptide is
cloned in a three-oligonucleotide
variation of the coiled-coil embodiment, thus allowing two different
randomizing regions to be
incorporated simultaneously. This embodiment utilizes non-palindromic BstXI
sites on the termini.
In a preferred embodiment, the presentation structure is a sequence that
contains generally two
cysteine residues, such that a disulfide bond may be formed, resulting in a
conformationally
constrained sequence. This embodiment is particularly preferred when secretory
targeting sequences
are used. As will be appreciated by those in the art, any number of random
sequences, with or without
spacer or linking sequences, may be flanked with cysteine residues. In other
embodiments, effective
presentation structures may be generated by the random regions themselves. For
example, the
random regions may be "doped" with cysteine residues which, under the
appropriate redox conditions,
may result in highly crosslinked structured conformations, similar to a
presentation structure. Similarly,
the randomization regions may be controlled to contain a certain number of
residues to confer fl-sheet
or a-helical structures.'

In a preferred embodiment, the fusion partner is a targeting sequence. As will
be appreciated by those
in the art, the localization of proteins within a cell is a simple method for
increasing effective
concentration and determining function. For example, RAF1 when localized to
the mitochondria)
membrane can inhibit the anti-apoptotic effect of BCL-2. Similarly, membrane
bound Sos induces Ras
mediated signaling in T-lymphocytes. These mechanisms are thought to rely on
the principle of limiting
the search space for ligands, that is to say, the localization of a protein to
the plasma membrane limits
the search for its ligand to that limited dimensional space near the membrane
as opposed to the three
dimensional space of the cytoplasm. Alternatively, the concentration of a
protein can also be simply
increased by nature of the localization. Shuttling the proteins into the
nucleus confines them to a
smaller space thereby increasing concentration. Finally, the ligand or target
may simply be localized
to a specific compartment, and inhibitors must be localized appropriately.

23


CA 02325597 2008-09-10
7724-54

Thus, suitable targeting sequences include, but are not limited to, binding
sequences capable of
causing binding of the expression product to a predetermined molecule or class
of molecules while
retaining bioactivity of the expression product, (for example by using enzyme
inhibitor or substrate
sequences to target a class of relevant enzymes); sequences signalling
selective degradation, of itself
or co-bound proteins; and signal sequences capable of constitutively
localizing the candidate
expression products to a predetermined cellular locale, including a)
subcellular locations such as the
Golgi, endoplasmic reticulum, nucleus, nucleoli, nuclear membrane,
mitochondria, chloroplast,
secretory vesicles, lysosome, and cellular membrane; and b) extracellular
locations via a secretory
signal. Particularly preferred is localization to either subcellular locations
or to the outside of the cell
via secretion.

In a preferred embodiment, the targeting sequence is a nuclear localization
signal (NLS). NLSs are
generally short, positively charged (basic) domains that serve to direct the
entire protein in which they
occur to the cell's nucleus. Numerous NLS amino acid sequences have been
reported including single
basic NLS's such as that of the SV40 (monkey virus) large T Antigen (Pro Lys
Lys Lys Arg Lys Val),
Kalderon (1984), et al., Cell, 39:499-509; the human retinoic acid receptor-f3
nuclear localization signal
(ARRRRP); NFKB p50 (EEVQRKRQKL; Ghosh et al., Cell 62:1019 (1990); NFKB p65
(EEKRKRTYE;
Nolan et al., Cell 64:961 (1991); and others (see for example Boulikas, J.
Cell. Biochem. 55(1):32-58
-(1994)) and double basic NLS's exemplified by that of the Xenopus
(African clawed toad) protein, nucleoplasmin (Ala Val Lys Arg Pro Ala Ala Thr
Lys Lys Ala Gly Gin Ala
Lys Lys Lys Lys Leu Asp), Dingwall, et al., Cell, 30:449-458, 1982 and
Dingwall, et at., J. Cell Biol.,
107:641-849; 1988). Numerous localization studies have demonstrated that NLSs
incorporated in
synthetic peptides or grafted onto reporter proteins not normally targeted to
the cell nucleus cause
these peptides and reporter proteins to be concentrated in the nucleus. See,
for example, Dingwall,
and Laskey, Ann, Rev: Cell Biol., 2:367-390, 1986; Bonnerot, et al., Proc.
Natl. Acad. Sci. USA,
84:6795-6799, 1987; Galileo, et al., Proc. Natl. Acad. Sci. USA, 87:458-462,
1990.

In a preferred embodiment, the targeting sequence is a membrane anchoring
signal sequence. This is =
particularly useful since many parasites and pathogens bind to the membrane,
in addition to the fact
that many intracellular events originate at the plasma membrane. Thus,
membrane-bound peptide
libraries are useful for both the identification of important elements in
these processes as well as for
the discovery of effective inhibitors. The invention provides methods for
presenting the randomized
expression product extracellularly or in the cytoplasmic space; see Fig 3. For
extracellular
presentation, a membrane anchoring region is provided at the carboxyl terminus
of the peptide
presentation structure. The randomized epression product region is expressed
on the cell surface and
presented to the extracellular space, such that it can bind to other surface
molecules (affecting their
function) or molecules present in the extracellular medium. The binding of
such molecules could

24


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
confer function on the cells expressing a peptide that binds the molecule. The
cytoplasmic region
could be neutral or could contain a domain that, when the extracellular
randomized expression product
region is bound, confers a function on the cells (activation of a kinase,
phosphatase, binding of other
cellular components to effect function). Similarly, the randomized expression
product-containing
region could be contained within a cytoplasmic region, and the transmembrane
region and
extracellular region remain constant or have a defined function.

Membrane-anchoring sequences are well known in the art and are based on the
genetic geometry of
mammalian transmembrane molecules. Peptides are inserted into the membrane
based on a signal
sequence (designated herein as ssTM) and require a hydrophobic transmembrane
domain (herein
TM). The transmembrane proteins are inserted into the membrane such that the
regions encoded 5'
of the transmembrane domain are extracellular and the sequences 3' become
intracellular. Of course,
if these transmembrane domains are placed 5' of the variable region, they will
serve to anchor it as an
intracellular domain, which may be desirable in some embodiments. ssTMs and
TMs are known for a
wide variety of membrane bound proteins, and these sequences may be used
accordingly, either as
pairs from a particular protein or with each component being taken from a
different protein, or
alternatively, the sequences may be synthetic, and derived entirely from
consensus as artificial
delivery domains.

As will be appreciated by those in the art, membrane-anchoring sequences,
including both ssTM and
TM, are known for a wide variety of proteins and any of these may be used.
Particularly preferred
membrane-anchoring sequences include, but are not limited to, those derived
from CD8, ICAM-2, IL-
8R, CD4 and LFA-1.

Useful sequences include sequences from: 1) class I integral membrane proteins
such as IL-2
receptor beta-chain (residues 1-26 are the signal sequence, 241-265 are the
transmembrane
residues; see Hatakeyama et al., Science 244:551 (1989) and von Heijne et al,
Eur. J. Biochem.
174:671 (1988)) and insulin receptor beta chain (residues 1-27 are the signal,
957-959 are the
transmembrane domain and 960-1382 are the cytoplasmic domain; see Hatakeyama,
supra, and
Ebina et al., Cell 40:747 (1985)); 2) class 11 integral membrane proteins such
as neutral endopeptidase
(residues 29-51 are the transmembrane domain, 2-28 are the cytoplasmic domain;
see Malfroy et al.,
Biochem. Biophys. Res. Commun. 144:59 (1987)); 3) type III proteins such as
human cytochrome
P450 NF25 (Hatakeyama, supra); and 4) type IV proteins such as human P-
glycoprotein
(Hatakeyama, supra). Particularly preferred are CD8 and ICAM-2. For example,
the signal
sequences from CD8 and ICAM-2 lie at the extreme 5' end of the transcript.
These consist of the
amino acids 1-32 in the case of CD8 (MASPLTRFLSLNLLLLGESILGSGEAKPQAP; Nakauchi
et al.,
PNAS USA 82:5126 (1985) and 1-21 in the case of ICAM-2 (MSSFGYRTLTVALFTLICCPG;
Staunton



CA 02325597 2008-09-10
77245-54

et al., Nature (London) 339:61 (1989)). These leader sequences deliver the
construct to the
membrane while the hydrophobic transmembrane domains, placed 3' of the random
candidate region,
serve to anchor the construct in the membrane. These transmembrane domains are
encompassed by
amino acids 145-195 from CD8
(PQRPEDCRPRGSVKGTGLDFACDIYIWAPLAGICVALLLSLIITLICYHSR; Nakauchi, supra) and 224-

256 from ICAM-2 (MVIIVTVVSVLLSLFVTSVLLCFIFGQHLRQQR; Staunton, supra).

Alternatively, membrane anchoring sequences include the GPI anchor, which
results in a covalent
bond between the molecule and the lipid bilayer via a glycosyl-
phosphatidylinositol bond for example
in DAF (PNKGSGTTSGTTRLLSGHTCFTLTGLLGTLVTMGLLT, with the bolded serine the site
of the
anchor; see Homans et al., Nature 333(6170):269-72 (1988), and Moran et at.,
J. Biol. Chem.
266:1250 (1991)). In order to do this, the GPI sequence from Thy-1 can be
cassetted 3' of the
variable region in place of a transmembrane sequence.

Similarly, myristylation sequences can serve as membrane anchoring sequences.
It is known that the
myristylation of c-src recruits it to the plasma membrane. This is a simple
and effective method of
membrane localization, given that the first 14 amino acids of the protein are
solely responsible for this
function: MGSSKSKPKDPSQR (see Cross et al., Mot. Cell. Biol. 4(9):1834 (1984);
Spencer et al.,
Science 262:1019-1024 (1993)). This motif has
already been shown to be effective in the localization of reporter genes and
can be used to anchor the
zeta chain of the TCR. This motif is placed 5' of the variable region in order
to localize the construct to
the plasma membrane. Other modifications such as palmitoylation can be used to
anchor constructs in
the plasma membrane; for example, palmitoylation sequences from the G protein-
coupled receptor
kinase GRK6 sequence (LLQRLFSRQDCCGNCSDSEEELPTRL, with the bold cysteines
being
palmitolyated; Stoffel et at., J. Biol. Chem 269:27791 (1994)); from rhodopsin
(KQFRNCMLTSLCCGKNPLGD; Barnstable et at., J. Mol. Neurosci. 5(3):207 (1994));
and the p21 H-
ras 1 protein (LNPPDESGPGCMSCKCVLS; Capon et at., Nature 302:33 (1983)).

In a preferred embodiment, the targeting sequence is a lysozomal targeting
sequence, including, for
example, a lysosomal degradation sequence such as Lamp-2 (KFERQ; Dice, Ann.
N.Y. Acad. Sci.
674:58 (1992); or lysosomal membrane sequences from Lamp-1
(MLIPIAGFFALAGLVLIVLIAYLIGRKRSHAGYQTI, Uthayakumar et at., Cell. Mol. Biol.
Res. 41:405
(1995)) or Lamp-2 (LVPIAVGAALAGVLILVLLAYFIGLKHHHAGYEQF, Konecki et Ia.,
Biochem.
Biophys. Res. Comm. 205:1-5 (1994), both of which show the transmembrane
domains in italics and
the cytoplasmic targeting signal underlined).

Alternatively, the targeting sequence may be a mitrochondrial localization
sequence, including
26


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
mitochondrial matrix sequences (e.g. yeast alcohol dehydrogenase III;
MLRTSSLFTRRVQPSLFSRNILRLQST; Schatz, Eur. J. Biochem. 165:1-6 (1987));
mitochondrial inner
membrane sequences (yeast cytochrome c oxidase subunit IV;
MLSLRQSIRFFKPATRTLCSSRYLL;
Schatz, supra); mitochondrial intermembrane space sequences (yeast cytochrome
c1;
MFSMLSKRWAQRTLSKSFYSTATGAASKSGKLTQKLVTAGVAAAGITASTLLYADSLTAEAMTA;
Schatz, supra) or mitochondrial outer membrane sequences (yeast 70 kD outer
membrane protein;
MKSFITRNKTAILATVAATGTAIGAYYYYNQLQQQQQRGKK; Schatz, supra).

The target sequences may also be endoplasmic reticulum sequences, including
the sequences from
calreticulin (KDEL; Pelham, Royal Society London Transactions B; 1-10 (1992))
or adenovirus E3/19K
protein (LYLSRRSFIDEKKMP; Jackson et al., EMBO J. 9:3153 (1990).

Furthermore, targeting sequences also include peroxisome sequences (for
example, the peroxisome
matrix sequence from Luciferase; SKL; Keller et al., PNAS USA 4:3264 (1987));
farnesylation
sequences (for example, P21 H-ras 1; LNPPDESGPGCMSCKCVLS, with the bold
cysteine
farnesylated; Capon, supra); geranylgeranylation sequences (for example,
protein rab-5A;
LTEPTQPTRNQCCSN, with the bold cysteines geranylgeranylated; Farnsworth, PNAS
USA 91:11963
(1994)); or destruction sequences (cyclin 61; RTALGDIGN; Klotzbucher et at.,
EMBO J. 1:3053
(1996)).
In a preferred embodiment, the targeting sequence is a secretory signal
sequence capable of effecting
the secretion of the candidate translation product. There are a large number
of known secretory signal
sequences which are placed 5' to the variable peptide region, and are cleaved
from the peptide region
to effect secretion into the extracellular space. Secretory signal sequences
and their transferability to
unrelated proteins are well known, e.g., Silhavy, et at. (1985) Microbiol.
Rev. 49, 398-418. This is
particularly useful to generate a peptide capable of binding to the surface
of, or affecting the
physiology of, a target cell that is other than the host cell, e.g., the cell
expressing the peptide. In a
preferred approach, a fusion product is configured to contain, in series,
secretion signal peptide-
presentation structure-randomized expression product region-presentation
structure. In this manner,
target cells grown in the vicinity of cells caused to express the library of
peptides, are bathed in
secreted peptide. Target cells exhibiting a physiological change in response
to the presence of a
peptide, e.g., by the peptide binding to a surface receptor or by being
internalized and binding to
intracellular targets, and the secreting cells are localized by any of a
variety of selection schemes and
the peptide causing the effect determined. Exemplary effects include variously
that of a designer
cytokine (i.e., a stem cell factor capable of causing hematopoietic stem cells
to divide and maintain
their totipotential), a factor causing cancer cells to undergo spontaneous
apoptosis, a factor that binds
to the cell surface of target cells and labels them specifically, etc.

27


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
Suitable secretory sequences are known, including signals from IL-2
(MYRMQLLSCIALSLALVTNS;
Villinger et at., J. Immunol. 155:3946 (1995)), growth hormone
(MATGSRTSLLLAFGLLCLPWLQEGSAE=; Roskam et at, Nucleic Acids Res. 7:30 (1979));
preproinsulin (MALWMRLLPLLALLALWGPDPAAAFVI; Bell et al., Nature 284:26
(1980)); and
influenza HA protein (MKAKLLVLLYAFVAGj? 1; Sekiwawa et al., PNAS 80:3563)),
with cleavage
between the non-underlined-underlined junction. A particularly preferred
secretory signal sequence is
the signal leader sequence from the secreted cytokine IL-4, which comprises
the first 24 amino acids
of IL-4 as follows: MGLTSQLLPPLFFLLACAGNFVHG.

In a preferred embodiment, the fusion partner is a rescue sequence. A rescue
sequence is a
sequence which may be used to purify or isolate either the candidate agent or
the nucleic acid
encoding it. Thus, for example, peptide rescue sequences include purification
sequences such as the
Hiss tag for use with Ni affinity columns and epitope tags for detection,
immunoprecipitation or FAGS
(fluoroscence-activated cell sorting). Suitable epitope tags include myc (for
use with the commercially
available 9E10 antibody), the BSP biotinylation target sequence of the
bacterial enzyme BirA, flu tags,
lacZ, and GST.

Alternatively, the rescue sequence may be a unique oligonucleotide sequence
which serves as a
probe target site to allow the quick and easy isolation of the retroviral
construct, via PCR, related
techniques, or hybridization.

In a preferred embodiment, the fusion partner is a stability sequence to
confer stability to the candidate
bioactive agent or the nucleic acid encoding it. Thus, for example, peptides
may be stabilized by the
incorporation of glycines after the initiation methionine (MG or MGGO), for
protection of the peptide to
ubiquitination as per Varshavsky's N-End Rule, thus conferring long half-life
in the cytoplasm.
Similarly, two prolines at the C-terminus impart peptides that are largely
resistant to carboxypeptidase
action. The presence of two glycines prior to the prolines impart both
flexibility and prevent structure
initiating events in the di-proline to be propagated into the candidate
peptide structure. Thus, preferred
stability sequences are as follows: MG(X)õ GGPP, where X is any amino acid and
n is an integer of at
least four.

In one embodiment, the fusion partner is a dimerization sequence. A
dimerization sequence allows
the non-covalent association of one random peptide to another random peptide,
with sufficient affinity
to remain associated under normal physiological conditions. This effectively
allows small libraries of
random peptides (for example, 104) to become large libraries if two peptides
per cell are generated
which then dimerize, to form an effective library of 10s (104 X 104). It also
allows the formation of
longer random peptides, if needed, or more structurally complex random peptide
molecules. The
28


CA 02325597 2000-10-16 _
WO 99/54494 PCTIUS99/08345
dimers may be homo- or heterodimers.

Dimerization sequences may be a single sequence that self-aggregates, or two
sequences, each of
which is generated in a different retroviral construct. That is, nucleic acids
encoding both a first
random peptide with dimerization sequence 1, and a second random peptide with
dimerization
sequence 2, such that upon introduction into a cell and expression of the
nucleic acid, dimerization
sequence 1 associates with dimerization sequence 2 to form a new random
peptide structure.
Suitable dimerization sequences will encompass a wide variety of sequences.
Any number of protein-
protein interaction sites are known. In addition, dimerization sequences may
also be elucidated using
standard methods such as the yeast two hybrid system, traditional biochemical
affinity binding studies,
or even using the present methods.

The fusion partners may be placed anywhere (i.e. N-terminal, C-terminal,
internal) in the structure as
the biology and activity permits.

In a preferred embodiment, the fusion partner includes a linker or tethering
sequence, as generally
described in PCT US 97/01019, that can allow the candidate agents to interact
with potential targets
unhindered. For example, when the candidate bioactive agent is a peptide,
useful linkers include
glycine-serine polymers (including, for example, (GS),,, (GSGGS)õ and (GGGS)n,
where n is an integer
of at least one), glycine-alanine polymers, alanine-serine polymers, and other
flexible linkers such as
the tether for the shaker potassium channel, and a large variety of other
flexible linkers, as will be
appreciated by those in the art. Glycine-serine polymers are preferred since
both of these amino acids
are relatively unstructured, and therefore may be able to serve as a neutral
tether between
components. Secondly, serine is hydrophilic and therefore able to solubilize
what could be a globular
glycine chain. Third, similar chains have been shown to be effective in
joining subunits of recombinant
proteins such as single chain antibodies.

In addition, the fusion partners, including presentation structures, may be
modified, randomized,
and/or matured to alter the presentation orientation of the randomized
expression product. For
example, determinants at the base of the loop may be modified to slightly
modify the internal loop
peptide tertiary structure, which maintaining the randomized amino acid
sequence.

In a preferred embodiment, combinations of fusion partners are used. Thus, for
example, any number
of combinations of presentation structures, targeting sequences, rescue
sequences, and stability
sequences may be used, with or without linker sequences.

29


CA 02325597 2008-09-10
7724%-54

Thus, candidate agents can include these components, and may then be used to
generate a library of
fragments, each containing a different random nucleotide sequence that may
encode a different
peptide. The ligation products are then transformed into bacteria, such as E.
co/i, and DNA is
prepared from the resulting library, as is generally outlined in Kitamura,
PNAS USA 92:9146-9150
(1995).

Delivery of the library DNA into a retroviral packaging system results in
conversion to infectious virus.
Suitable retroviral packaging system cell lines include, but are not limited
to, the Bing and BOSC23 cell
lines described in WO 94/19478; Soneoka et al., Nucleic Acid Res. 23(4):628
(1995); Finer et al.,
Blood 83:43 (1994); Pheonix packaging lines such as PhiNX-eco and PhiNX-ampho,
described below;
292T + gag-pol and retrovirus envelope; PA317; and cell lines outlined in
Markowitz et al., Virology
167:400 (1988), Markowitz et al., J. Virol. 62:1120 (1988), Li et at, PNAS USA
93:11658 (1996),
Kinsella et al., Human Gene Therapy 7:1405 (1996).
Preferred systems include PhiNX-eco and PhiNX-ampho or similar cell lines,
disclosed in PCT
US97/01019.

When the cells are not replicating, other viral vectors may be used, including
adenoviral vectors, feline
immunoviral (FIV) vectors, etc.

In a preferred embodiment, when the candidate agent is introduced to the cells
using a viral vector, the
candidate peptide agent is linked to a detectable molecule, and the methods of
the invention include at
least one expression assay. An expression assay is an assay that allows the
determination of
whether a candidate bioactive agent has been expressed, i.e. whether a
candidate peptide agent is
present in the cell. Thus, by linking the expression of a candidate agent to
the expression of a
detectable molecule such as a label, the presence or absence of the candidate
peptide agent may be
determined. Accordingly, in this embodiment, the candidate agent is operably
linked to a detectable
molecule. Generally, this is done by creating a fusion nucleic acid. The
fusion nucleic acid comprises
a first nucleic acid encoding the candidate bioactive agent (which can include
fusion partners, as
outlined above), and a second nucleic acid encoding a detectable molecule. The
terms "first" and
"second" are not meant to confer an orientation of the sequences with respect
to 5'-3' orientation of the
fusion nucleic acid. For example, assuming a 5'-3' orientation of the fusion
sequence, the first nucleic
acid may be located either 5' to the second nucleic acid, or 3' to the second
nucleic acid. Preferred
detectable molecules in this embodiment include, but are not limited to,
fluorescent proteins, including
GFP, YFP, BFP and RFP, with the former being especially preferred.
In general, the candidate agents are added to the cells (either
extracellularly or intracellularly, as
outlined above) under reaction conditions that favor agent-target
interactions. Generally, this will be


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
physiological conditions. Incubations may be performed at any temperature
which facilitates optimal
activity, typically between 4 and 40 C. Incubation periods are selected for
optimum activity, but may
also be optimized to facilitate rapid high through put screening. Typically
between 0.1 and 1 hour will
be sufficient. Excess reagent is generally removed or washed away.
A variety of other reagents may be included in the assays. These include
reagents like salts, neutral
proteins, e.g. albumin, detergents, etc which may be used to facilitate
optimal protein-protein binding
and/or reduce non-specific or background interactions. Also reagents that
otherwise improve the
efficiency of the assay, such as protease inhibitors, nuclease inhibitors,
anti-microbial agents, etc.,
may be used. The mixture of components may be added in any order that provides
for detection.
Washing or rinsing the cells will be done as will be appreciated by those in
the art at different times,
and may include the use of filtration and centrifugation. When second labeling
moieties (also referred
to herein as "secondary labels") are used, they are preferably added after
excess non-bound target
molecules are removed, in order to reduce non-specific binding; however, under
some circumstances,
all the components may be added simultaneously.

In a preferred embodiment, the cells are sorted using fluorescent-activated
cell sorting (FACS). In the
invention herein, cell cycle regulation is evaluated by multiple parameters
which results in reduced
background and greater specificity. In contrast, FACS has been used in the
past to evaluate two
different or unrelated characteristics at the same time which identifies cells
having those two
characteristics, but does not reduce the background for the combined
characteristics.

Thus, the cells are sorted or enriched in a FACS on the basis of one or more
of the assays, including a
cell viability assay, a proliferation assay, a cell phase assay, and (when
candidate agents are
expressed with detectable moieties) an expression assay. The results from one
or more of these
assays are compared to cells that were not exposed to the candidate bioactive
agent, or to the same
cells prior to introduction of the candidate agent. Alterations in these
results can indicate that said
agent modulates cell cycle regulation.

A strength of the present invention is that a library of candidate agents may
be tested in a library of
cells, because the present methods allow single cell sorting, with extremely
high specificity, such that
very rare events may be detected. The use of multiple laser paths allows sort
accuracy of 1 in 106
with better than 70% accuracy.

In addition, the present invention can, in addition to the identification of
multiple cell cycle regulation
properties, be combined with the identification of other cellular
characteristics. For example,
parameters of general cellular health can be determined and selected for by
using i.e., dye Indo-1

31


CA 02325597 2000-10-16

WO 99/54494 PCTIUS99/08345
indicating a calcium response. Other cellular parameters which are routinely
identified by the skilled
artisan include but are not limited to: cell size, cell shape, redox state,
DNA content, nucleic acid
sequence, chromatin structure, RNA content, total protein, antigens, lipids,
surface proteins,
intracellular receptors, oxidative metabolism, DNA synthesis and degradation
and intracellular pH.
In a preferred embodiment, each of the measurements is determined
simultaneously from an
individual cell as it passes through the beam paths of multiple lasers.
Alternatively, the measurements
are done sequentially. By using more than one parameter to detect cell cycle
regulation or alterations
in cell cycle regulation, background is reduced and specificity is increased.
The cells meeting the
parameters of the desired properties can be physically sorted from cells not
meeting the desired
parameters or they can be identified by their percentage in the cell
population.

In general, K0 s of < 1 pM are preferred, to allow for retention of binding in
the presence of the shear
forces present in FACS sorting. In a preferred embodiment, the cells are
sorted at very high speeds,
for example greater than about 5,000 sorting events per sec, with greater than
about 10,000 sorting
events per sec being preferred, and greater than about 25,000 sorting events
per second being
particularly preferred, with speeds of greater than about 50,000 to 100,000
being especially preferred.
Cells processed for stimulation and staining are generally taken up in buffer
and filtered prior to
cytometry. Cells can be analyzed using a FACSCAN (Becton Dickinson Inc., laser
line 488nm) or a
Mo-Flo (Cytomation, Inc., laser lines 350nM broadband (UV), 488nm, and 647nm)
Cytometer. Cells
are sorted, if desired, using the Mo-Flo.

Wherein the cells are analyzed by microscopy, cells post stimulation or
staining are generally mounted
onto glass slides and coverslipped; these are directly visualized by
brightfield and fluorescence
microscopy on an inverted microscope (i.e., TE300, Nikon) using standard BFP,
FITC, or TRITC (for
example) filter sets. Images can also be obtained using an inverted confocal
scanning microscope
(Zeiss, Inc,, Bio-Rad, Inc.) using standard FITC and TRITC (for example)
filter sets.

The sorting results in a population of cells having the desired properties. In
a preferred embodiment,
the parameters are set to identify at least one candidate bioactive agent that
modulates cell cycle
regulation.

In a preferred embodiment, the bioactive agent is characterized. This will
proceed as will be
appreciated by those in the art, and generally includes an analysis of the
structure, identity, binding
affinity and function of the agent. Generally, once identified, the bioactive
agent is resynthesized and
combined with the target cell to verify the cell cycle regulation modulation
under various conditions

32


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
and in the presence or absence of other various agents. The bioactive can be
prepared in a
therapeutically effective amount to modulate cell cycle regulation and
combined with a suitable
pharmaceutical carrier.

In a preferred embodiment, the cell populations can be subjected to various
experimental conditions,
with and without the candidate agents. Changes in conditions include but are
not limited to changes in
pH, temperature, buffer or salt concentration, etc. In a preferred embodiment,
the pH is changed,
generally by increasing or decreasing the pH, usually by from about 0.5 to
about 3 pH units.
Alternatively, the temperature is altered, with increases or decreases of from
about 5 C to about 30 C
being preferred. Similarly, the salt concentration may be modified, with
increases or decreases of
from about 0.1 M to about 2 M being preferred.

It is understood by the skilled artisan that the steps of the assays provided
herein can vary in order. It
is also understood, however, that while various options (of compounds,
properties selected or order of
steps) are provided herein, the options are also each provided individually,
and can each be
individually segregated from the other options provided herein. Moreover,
steps which are obvious
and known in the art that will increase the sensitivity of the assay are
intended to be within the scope
of this invention. For example, there may be additionally washing steps, or
segregation, isolation
steps. Moreover, it is understood that in some cases detection is in the
cells, but can also take place
in the media, or vice versa.

In a preferred embodiment, the cellular phenotype is exocytosis, and the
methods and compositions of
the invention are directed to the detection of alterations in exocytosis,
again using a FACS machine.
There are a number of parameters that may be evaluated or assayed to allow the
detection of
alterations in exocytotic pathways, including, but not limited to, light
scattering, fluorescent dye uptake,
fluorescent dye release, granule exposure, surface granule enzyme activity,
and the quantity of
granule specific proteins. By assaying or measuring one or more of these
parameters, it is possible to
detect not only alterations in exocytosis, but alterations of different steps
of the exocytotic pathway. In
addition, multiparameter analysis also reduces the background, or "false
positives", that are detected.
In this manner, rapid, accurate screening of candidate agents may be performed
to identify agents that
modulate exocytosis.

In a preferred embodiment, the invention provides methods for screening for
alterations in exocytosis
of a population of cells. By "alteration" or "modulation" in the context of
exocytosis is meant a
decrease or an increase in the amount of exocytosis in one cell compared to
another cell or in the
same cell under different conditions. The measurements can be determined
wherein all of the
conditions are the same for each measurement, or under various conditions,
with or without bioactive

33


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
agents, or at different stages of the exocytic process. For example, a
measurement of exocytosis can
be determined in a cell population wherein a candidate bioactive agent is
present and wherein the
candidate bioactive agent is absent. In another example, the measurements of
exocytosis are
determined wherein the condition or environment of the populations of cells
differ from one another.
For example, the cells may be evaluated in the presence or absence of
physiological signals, such as
exocytic inducers (i.e, Ca", ionomycin, etc.), hormones, antibodies, peptides,
antigens, cytokines,
growth factors, action potentials, or other cells (i.e. cell-cell contacts).
In another example, the
measurements of exocytosis are determined at different-stages of the exocytic
process. In yet another
example, the measurements of exocytosis are taken wherein the conditions are
the same, and the
alterations are between one cell or cell population and another cell or cell
population.

By a "population of cells" herein is meant a sample of cells as defined above.
In this embodiment, the
cells are preferably (but not required) to be rapidly growing, retrovirally
infectable, and compatible with
dyes and antibodies. Preferred cell types for use in this embodiment, include,
but are not limited to,
mast cells, neurons, adrenal chromaffin cells, basophils, endocrine cells
including pancreatic p-cells,
pancreatic acinar cells including exocrine cells, neutrophils, monocytes,
lymphocytes, mammary cells,
sperm, egg cells and PMN leukocytes, endothelial cells, adipocytes, and muscle
cells.

The exocytotic methods comprise sorting the cells in a FACS machine by
assaying for alterations in at
least three of the properties selected from the group consisting of light
scattering, fluorescent dye
uptake, fluorescent dye release, granule exposure, surface granule enzyme
activity, and the quantity
of granule specific proteins. In a preferred embodiment, each of the
measurements is determined
simultaneously from an individual cell as it passes through the beam paths of
multiple lasers.
Alternatively, the measurements are done sequentially. By using more than one
parameter to detect
exocytosis or alterations in exocytosis, background is reduced and specificity
is increased. The cells
meeting the parameters of the desired properties can be physically sorted from
cells not meeting the
desired parameters or they can be identified by their percentage in the cell
population.

In a preferred embodiment, changes in light scattering are assayed to
determine alterations in
exocytosis in a population of cells. When viewed in the FACS, cells have
particular characteristics as
measured by their forward and 90 degree (side) light scatter properties. These
scatter properties
represent the size, shape and granule-content of the cells. Upon activation of
the cells with a pro-
exocytic stimulus, both the forward and side scatter properties of the cells
changes considerably.
These properties account for two parameters to be measured as a readout for
the exocytic event.
These properties change in proportion to the extent of exocytosis of the cells
and depend on the time
course of the exocytic events as well. Alterations in the intensity of light
scattering, or the cell-
refractive index indicate alterations in exocytosis either in the same cell at
different times, or compared

34


CA 02325597 2008-09-10
7724-54

to the same cell under different conditions or with candidate bioactive agents
present or absent, or
compared to different cells or cell populations.

In one embodiment provided herein, a cell population is combined with an agent
which is known to
stimulate exocytosis and the light scattering properties are determined. Cells
having light scattering
properties indicating the desirable exocytic activity can be identified and/or
sorted. Exocytic activity as
used herein includes lack of activity. In a preferred embodiment, candidate
bioactive agents are
combined with the cell population prior to or with the exocytic stimulus, as
is more fully outlined below.
In this embodiment, where light scattering properties differ as between a) a
cell population combined
with a known exocytic stimulus and a candidate bioactive agent, and b) a cell
population combined
with a known exocytic stimulus wherein the candidate bioactive agent is
absent, it can be determined
that the candidate bioactive agent modulates exocytosis. = It may also be
desirable in some cases to
include an inhibitor of exocytosis or to exclude the exocytic stimulus to
identify bioactive agents which
induce exocytosis. Preferably, light scattering properties are measured in
combination with at least
one, and preferably two other properties which indicate exocytosis activity.
General methodologies for
light scattering measurements are further described in Perretti, et al., J.
Pharmacol. Methods,
23(3):187-194 (1990) and Hide et al., J. Cell Biol., 123(3):585-593 (1993).
In general, changes of at least about 5% from baseline are preferred, with at
least about
25% being more preferred, at least about 50% being particularly preferred, and
at least about 75 to
100% being especially preferred. Baseline in this case generally means the
light scatter properties of
the cells prior to exocytotic stimulation. In each case provided herein, the
baseline may also be set for
any control parameter. For example, the baseline may be set at the exocytosis
measurement of a
particular cell, a similar cell under different conditions, or at a particular
time point during exocytosis.

In another preferred embodiment, changes in fluorescent dye uptake are
evaluated. Preferred
fluorescent dyes include styryl dyes, which indicate exocytosis activity in
relation to endocytosis,
sometimes referred to as coupled endocytosis. The theory behind coupled
endocytosis is that cells
undergoing exocytosis must also undergo endocytosis in order to maintain cell
volume and membrane
integrity. Thus, upon exocytic stimulation, endocytosis is also increased,
providing an indirect
measurement of exocytosis by quantifying the amount of styryl dye uptake.

In an embodiment provided herein, the cells are bathed in a solution of styryl
dye and stimulated with a
pro-exocytic stimulus and the dye is quantitated. Preferably, after exocytic
stimulation, the cells are
spun down, aspirated and resuspended in fresh buffer. In a preferred
embodiment, a candidate
bioactive agent is combined with the cells as described herein. In some cases,
the candidate
bioactive agent can be combined with the cells with an inhibitor of exocytosis
or without the pro-
exocytic stimulus. Preferably, a pro-exocytic stimulus is added to the cell
population which results in a



CA 02325597 2008-09-10
77245-54

dramatic increase in the fluorescence signal of the dye. The increased cell
associated signal is due to
coupled endocytosis of the styryl dye and is proportional to the exocytic
response in both time and
intensity. Conversely, the signal is not increased wherein exocytosis is
inhibited or is not induced.
Alterations are determined by measuring the fluorescence at either different
time points or in different
cell populations, and comparing the determinations to one another or to
standards. In general,
changes of at least about 50% from baseline are preferred, with changes of at
least about 75%-100%
being more preferred, changes of at least about 250% being particularly
preferred, and changes of at
least about 1000-2000% being especially preferred. Baseline in this case means
the styryl dye uptake
of cells prior to exocytic stimulation.
Preferred styryl dyes include, but are not limited to FM1-43, FM4-64; FM 14-
68, FM2-10, FM4-84,
FMI-84, FM14-27, FM14-29, FM3-25, FM3-14, FM5-55, RH414, FM6-55, FM10-75, FM1-
81, FM9-49,
FM4-95, FM4-59, FM9-40, and combinations thereof. Preferred dyes such as FM1-
43 are only weakly
fluorescent in water but very fluorescent when associated with a membrane,
such that dye uptake is
readily discernable. Suitable dyes are available commercially, i.e., Molecular
Probes, Inc., of Eugene,
Oregon, "Handbook of Fluorescent Probes and Research Chemicals", 6th Edition,
1996, particularly,
Chapter 17, and more particularly, Section 2 of Chapter 17, (including
referenced related chapter).
Preferably, the dyes are provided in a solution wherein the
dye concentration is about 25 to 1000- 5000 nM, with from about 50 to about
1000 nM being preferred,
-20 and from about 50 to 250 being particularly preferred. The use of styryl
dyes is further described in
Betz, et at., Current Opinion in Neurobiology, 6:365-371 (1996).
Preferably, fluorescent dye uptake is measured in combination with at least
one, and preferably two
other indicators of exocytosis activity.

In another preferred embodiment, changes in fluorescent dye release are
evaluated. The present
invention is in part directed to the discovery that low pH concentration dyes,
which are normally used
to stain lysozomes, also low pH stain exocytic granules. Generally, these dyes
can be taken up by the
cells passively and concentrate in granules; however, the cells can be induced
to take up the dye, i.e.,
by coupled endocytosis. In a preferred embodiment, a cell population is bathed
in a low pH
concentration dye such that the dye is taken up by the cells. The cells are
preferably washed. The
cells can be exposed to a pro-exocytic stimulus and/or inhibitor. In a
preferred embodiment, a
candidate bioactive agent is combined with the cell population and preferably,
the pro-exocytic
stimulus. Fluorescence is evaluated. Changes in fluorescent dye release
between cells or at different
time points in the same cell indicate alterations in exocytosis. Preferably,
the alterations are between
cells, and most preferably, between cells having different bioactive agents
added thereto. Changes of
at least about 5% from baseline are preferred, with at least about 25% being
more preferred, at least
about 50% being particularly preferred and at least about 100% being
especially preferred. Baseline in
36


CA 02325597 2008-09-10
7724-54

this case means the amount of dye in the cells prior to stimulation.

In this embodiment, low pH concentration dyes are preferred. Such low pH
concentration dyes include
but are not limited to acridine orange, LYSOTRACKERTM red, LYSOTRACKERTM
green, and
LYSOTRACKERTM blue. Such dyes are commercially available, i.e., from Molecular
Probes, supra,
particularly including Chapter 17, Section 4 of Chapter 17, and referenced
"related chapters", i.e.,
Chapter 23. In preferred embodiments, the dyes are administered in a solution
wherein the dye is a
concentration of about 50 nM to about 25 pM, with from about 5 pM to about 25
pM being preferred,
and from about 1 to 5 pM being particularly preferred. The use of low pH
concentration dyes is
generally described (in regards to lysozome studies) in Haller, et al., Cell
Calcium, 19(2):157-165
(1996).

In an alternative embodiment wherein changes in fluorescent dye release are
evaluated, the
fluorescence released into the supernatant is evaluated. In this embodiment,
either styryl dyes, which
reversibly label endocytosed membranes, or low pH concentration dyes are used.
In this embodiment,
a cell population is bathed in dye such that the dye is taken up into the
cells passively or by induction.
The cells are then preferably washed. The cells can be exposed to a pro-
exocytic stimulus and/or
inhibitor, and optionally, a candidate bioactive agent. The cells which are
exposed to a pro-exocytic
stimulus will release the dye into the extracellular medium. The fluorescence
in the medium can be
measured or detected. This process is sometimes referred to as destaining the
cells. Optionally, an
agent for improving and facilitating the detection of the dye in the medium
can be added. For
example, micelle-forming detergents such as 3-[(3-
cholamidopropyl)dimethylammonio] -1-
propanesulfonate (CHAPS) increase the fluorescence and thereby allow detection
of small amounts of
exocytosis activity. Changes in the release of dye will indicate alterations
in exocytosis in the same
cell, between cells, and most preferably, between cells having different
bioactive agents added
thereto. In general, changes of at least about 5% from baseline are preferred,
with at least about 25%
being more preferred, with at least about 50% being particularly preferred and
at least about 100%
being especially preferred. Baseline in this case means the release of dye
prior to exocytotic stimulus.
Preferably, dye release when measured in the media is combined with the
evaluation of at least one
other exocytosis indicator.

In a preferred embodiment, changes in granule exposure are determined. The
granules are exposed
to the media during exocytosis, i.e., the granules fuse with the cell membrane
and expose/release
their contents. Therefore, granule exposure is indicative of exocytic
activity, and its absence is
indicative that exocytosis has not been induced, or has been inhibited.
Preferably, granule exposure
is detected by a detectable agent which specifically bind to granules. An
example of a detectable
agent used herein is annexin V, a member of a protein family which displays
specific binding to

37


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
phospholipid (phosphatidylserine) in a divalent ion dependent manner. This
protein has been widely
used for the measurement of apoptosis (programmed cell death) as cell surface
exposure of
phosphatidylserine is a hallmark early signal of this process. Surprisingly,
it has been determined
herein that annexin V specifically binds to exocytic granules when they are
exposed at the cell surface
during the secretory process; granules internal to the cell are unlabeled.
This property of annexin V is
used herein to create a single exocytosis assay based on its exocytosis
dependent binding. Upon
exocytic stimulation of cells, the cells show an increase in annexin binding
and fluorescent signal in
proportion in both time and intensity to the exocytic response.

In this embodiment, annexin is labelled, either directly or indirectly, and
combined with a cell
population. Annexin is commercially available, i.e., from PharMingen, San
Diego, California, or Caltag
Laboratories, Millbrae, California. Preferably, the annexin is provided in a
solution wherein the
annexin is in a concentration of about 100 ng/ml to about 500 ng/ml, more
preferably, about 500 ng/ml
to about 1 pg/ml, and most preferably, from about 1 pg/ml to about 5 pg/ml. In
a preferred
embodiment, the annexin is directly labelled; for example, annexin may be
labelled with a
fluorochrome such as fluorecein isothiocyanate (FITC), Alexa dyes, TRITC,
AMCA, APC, tri-color, Cy-
5, and others known in the art or commercially available. In an alternate
preferred embodiment, the
annexin is labelled with a first label, such as a hapten such as biotin, and a
secondary fluorescent
label is used, such as fluorescent streptavidin. Other first and second
labelling pairs can be used as
will be appreciated by those in the art.

In the preferred embodiment, the cells are subjected to conditions that
normally cause exocytosis.
Optionally, a candidate bioactive agent is added to the cells. In some cases,
it may be desirable to
include an inhibitor of exocytosis to determine whether the candidate agent
can reverse the inhibition,
or to add the candidate bioactive agent without an exocytic stimulus to
determine whether the agent
induces exocytosis. The cells are preferably washed and fluorescence is
detected in the microscope
or on the flowcytometer. Alterations in the detection of annexin binding
indicates alterations in
exocytosis in the same cell, or between different cells, with or with the same
conditions and/or agents
combined therewith. In general, changes of at least about 25% from baseline
are preferred, with at
least about 50% being more preferred, at least about 100 being particularly
preferred and at least
about 500% being especially preferred. Baseline in this case means the amount
of annexin binding
prior to exocytic stimulation.

In another preferred embodiment, granule exposure is detected by a cationic
dye such as berberine or
ruthenium red. Such cationic dyes specifically stain secreting granules. Thus,
when exocytosis
occurs, and secreting granules are exposed at the cell surface, an increase in
fluorescence can be
detected. In a preferred embodiment, the cationic dye is combined with a cell
population in the

38


CA 02325597 2008-09-10
7724-54

presence or absence of an exocytic stimulus and/or inhibitor, and optionally,
in the presence or
absence of a candidate bioactive agent. In a particularly preferred
embodiment, the berberine is
combined with a cell and an exocytic stimulus and a candidate bioactive agent
to determine whether
the candidate bioactive agent can modulate the exocytic activity. Preferably,
the cells are washed and
then fluorescence is determined. In preferred embodiments, cationic dye
evaluation is combined with
evaluation of at least one other indicator of exocytosis. The dye is combined
with the cells as is known
in the art. General methodologies describing berberine are described in Berlin
and Enerback, Int. Arch.
Allergy Appl. Immunol., 73(3):256-262 (1984). In general, changes
of at least about 5% from baseline are preferred, with at least about 25%
being more preferred, at
least about 50% being particularly preferred, and at least about 100% being
especially preferred.
Baseline in this case means the amount of dye binding prior to stimulation.

Similarly, Con A-FITC can be used, as it binds to the carbohydrate on granule
proteins, in a manner
similar to those outlined herein.
In another preferred embodiment, changes in surface granule enzyme activity is
determined.
Secretory granules contain enzymes such as proteases and glycosidases which
are released as part
of the exocytic process. Frequently, these enzymes are inactive within the
granule, due to the low pH,
but upon exposure to the extracellular media at physiological pH, they become
activated. These
enzyme activities can be measured using chromogenic or fluorogenic substrates
as components of
the extracellular media. This allows detection of exocytic cells in varying
approaches.

In one embodiment, sometimes called herein the population based enzyme assay,
the generation of
signal via cleavage of a chromogenic or fluorogenic substrate can be
quantified in the media. That is,
the amount of detectable reaction product in the media is related to the
amount of enzyme present,
and thus to the amount of exocytosis. In this embodiment, it is the media, not
the cells, that becomes
detectable.

In a preferred embodiment, cells are subjected to an exocytic stimulus, and
optionally, a candidate
bioactive agent. The chromogenic or fluorogenic substrate is added to the
media, and changes in the
signal are evaluated, as the enzymes cleave the extracellular substrates.

In an alternate preferred embodiment, sometimes called herein "in. situ
enzymology assay", fluorogenic
substrates that precipitate upon cleavage are used. That is, upon exocytosis a
considerable amount
of enzyme activity remains cell/granule associated and can be visualized using
fluorescent substrates
which precipitate at the site of activity. For example, substrates for
glucuronidase, such as ELF-97
glucuronide, precipitate on exocytosing cells, but not resting cells, and thus
the cells can show

39


CA 02325597 2008-09-10
7724-54

increased fluorescence. The fluorescence is a direct measurement of exocytosis
and is pH dependent
reflecting the pH optima of the exocytosed enzyme. This method also provides a
method of
distinguishing different subtypes of granules based on their enzyme profile.

In a preferred embodiment, the cell population is subjected to an exocytic
stimulus and then incubated
with a detectable substrate. A candidate bioactive agent is optionally added.
The cells are washed
and then viewed in the microscope or flowcytometer.

Preferred granule enzymes include but are not limited to chymase, tryptase,
arylsulfatase A, beta-
hexosaminidase, beta-glucuronidase, and beta-D-galactosidase. Substrates
include ELF-97
glucuronide, N-acetyl beta-D glucoronide, ELF-97 coupled to peptides, etc.,
many of which are
commercially available, i.e., from Molecular Probes, supra, particular Chapter
10, more particularly
Section 2 of Chapter 10, and referenced "related chapters".

By detectable substrate is meant that the substrate comprises a fluorescent
molecule as further
described herein, or can be detected with a fluorescent molecule specific for
the substrate or cleaved
substrate, i.e., a fluorescent antibody. In a preferred embodiment, the
substrate comprises a
detectable molecule formed of two fluorescent proteins, i.e., blue and green
fluorescent protein (BFP
and GFP), and other similar molecules. As is known in the art, constructs of
GFP and BFG that hold
these two proteins in close proximity allow fluorescence resonance energy
transfer (FRET). That is,
the excitation spectra of the GFP overlaps the emission spectra of the BFP.
Accordingly, exciting the
BFP results in GFP emission. If a protease cleavage site is engineered between
the GFP and BFP to
form a "FRET construct", upon exposure of the FRET construct to an active
protease which cleaves
the construct, the GFP and BFP molecules separate. Thus, exciting the GFP
results in BFP emission
and loss of BFP emission.

Preferably, the protease dependent cleavage site inserted between two
fluoroscent proteins of the
FRET construct is specific for a granule specific enzyme. Thus, the FRET
construct can be used for
detecting granule specific proteases specific for the cleavage site of the
FRET construct. In this
embodiment, the protease substrate that is combined with'the cells or media
includes the FRET
construct. The FRET system allows for detection of the detectable molecule in
its cleaved and
uncleaved state, and distinguishes between the two. The system is further
described in Xu et al.,
Nucleic Acid Res. 26(8):2034 (1998); and Miyawaki et al., Nature 388(6645):882-
887 (1997).
.35
The amount of substrate added to the cells or media will depend in part on the
enzyme's specific
activity and the substrate itself, but generally is about 250 nM to about 1
mM, from about 1 pM to


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
about 100 pM being preferred, and from about 1 pM to about 10 pM being
particularly preferred. In
general, changes of at least about 5% from baseline are preferred, with at
least about 25% being
preferred, at least about 100% being particularly preferred and at least about
1000% being especially
preferred. Baseline in this case means the amount of substrate cleavage prior
to induction of
exocytosis.

In a preferred embodiment, changes in the quantity of granule specific
proteins are determined.
Secretory granules contain proteins which are specifically targeted to the
granule compartment due to
specific properties of these proteins. Upon exocytic induction, the granule
specific proteins are
exposed to the surface and detected.

In a preferred embodiment, detectable granule specific proteins are combined
with a population of
cells and subjected to conditions known to induce exocytosis. Optionally, a
bioactive candidate is
combined with the cell population and detectable granule specific protein and
the granule specific
protein is detected. Granule specific proteins include but are not limited to
VAMP and synaptotagmin.
Also included within the definition of granule specific proteins are the
mediators released during
exocytosis, including, but not limited to, serotonin, histamine, heparin,
hormones, etc.

The quantification of the granule proteins may be done in several ways. in one
embodiment, labelled
antibodies, (such as fluoroscent antibodies), to granule specific proteins are
used. In another
embodiment, the cells are engineered to contain fusion proteins comprising a
granule protein and a
detectable molecule. In a preferred embodiment, a detectable molecule is added
to the cells for
detection. For example, either directly or indirectly labelled antibodies can
be used. A preferred
embodiment uses a first labelled antibody, with fluorescent labels preferred.
Another embodiment
uses a first and second label, for example, a labelled secondary antibody.
Generally, this embodiment
may use any agent that will specifically bind to the granule protein or
compound that can be either
directly or indirectly labelled.

In a preferred embodiment the labels are engineered into the cells. For
example, recombinant
proteins are introduced to the cell population which are fusion proteins of a
granule specific protein
and a detectable molecule. This is generally done by transforming the cells
with a fusion nucleic acid
encoding a fusion protein comprising a granule specific protein and a
detectable molecule. This is
generally done as is known in the art, and will depend on the cell type.
Generally, for mammalian
cells, retroviral vectors and methods are preferred.
The fusion proteins are constructed by methods known in the art. For example,
the nucleic acids
encoding the granule specific protein is ligated with a nucleic acid encoding
a detectable molecule. By
41


CA 02325597 2000-10-16

WO 99/54494 PCT/US"/08345
detectable molecule herein is meant a molecule that allows a cell or compound
comprising the
detectable molecule to be distinguished from one that does not contain it,
i.e., an epitope, sometimes
called an antigen TAG, or a fluorescent molecule. Preferred fluorescent
molecules include but are not
limited to GFP, BFP, YFP, enzymes including luciferase and 0-galactosidase.
These constructs can be
made in such a way so that upon exocytosis an epitope, internal to the
granule, is exposed at the cell
surface and can then be detected. The epitope is preferably any detectable
peptide which is not
generally found on the cytoplasmic membrane, although in some instances, if
the epitope is one
normally found on the cells, increases may be detected, although this is
generally not preferred.

In a preferred embodiment, the cell population containing the fusion protein
or detectable granule
specific protein is subjected to exocytic conditions. Optionally, a candidate
bioactive agent and/or
exocytic inhibitor is included. Preferably, the cells are washed. Fluorescence
is detected on the cells.
In general, changes of at least about 5% from baseline are preferred, with at
least about 25% being
more preferred, at least about 50% being particularly preferred and at least
about 100% being
especially preferred. Generally, baseline in this case means amount of
fluorescence prior to exocytic
stimulus.

In the invention herein, the same characteristic of exocytosis is evaluated by
multiple parameters
which results in reduced background and greater specificity. In contrast, FACS
has been used in the
past to evaluate two different or unrelated characteristics at the same time
which identifies cells having
those two characteristics, but does not reduce the background for the combined
characteristics. The
present invention can, however, in addition to the identification of multiple
exocytosis properties, be
combined with the identification of other cellular parameters, as outlined
above.

In a preferred embodiment, the cells are subjected to conditions that normally
cause exocytosis. Pro-
exocytic agents include ionomycin, Ca**, ionophores (Ionomycin, AZ3187),
compound 48/80,
substance P, complement C3a/C5a, trypsin, tryptase, insulin, interleukin-3,
specific IgE, allergen, anti-
IgE, or anti-IgG receptor antibodies. These are provided at concentrations
depending on the
compound as is known in the art, ranging from 1 picomolar to 10 NM, generally.
In some cases, it
may be desirable to combine the cells with agents which inhibit exocytosis.
Exocytosis inhibitors
include but are not limited to Wortmannin, and Genestein, and others known in
the art.

In a preferred embodiment, the methods are used to screen candidate bioactive
agents for the ability
to modulate exocytosis. The candidate bioactive agents may be combined with
the cell population
before, during or after exocytosis is stimulated, preferably before. In some
instances, it may be
desirable to determine the effect of the candidate bioactive agent, also
referred to as "candidate
agents" herein, on the cell wherein exocytosis is not induced or wherein
exocytosis is inhibited. The
42


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
candidate bioactive agent can be added to the cell population exogenously or
can be introduced into
the cells as described further herein.

In a preferred embodiment, as above for cell cycle assays, a library of
different candidate bioactive
agents are used.

As above, the candidate bioactive agents are combined or added to a cell or
population of cells; again,
as outlined above, preferred embodiments utilize nucleic acid candidate agents
and fusion partners;
and preferably retroviral constructs.
Wherein the candidate agents are nucleic acids, methods known in the art such
as calcium phosphate,
electroporation, and injection may be used to introduce these to the cells.
The exocytic stimulus is
generally combined with the cells under physiological conditions. Incubations
may be performed at
any temperature which facilitates optimal activity, typically between 4 and 40
C. Incubation periods
are selected for optimum activity, but may also be optimized to facilitate
rapid high through put
screening.

As above, a variety of other reagents may be included in the assays, and the
cells are sorted as
above. The sorting results in a population of cells having the desired
exocytic properties. In a
preferred embodiment, the parameters are set to identify at least one
candidate bioactive agent that
modulates exocytosis.

In a preferred embodiment, the bioactive agent is characterized. This will
proceed as will be
appreciated by those in the art, and generally includes an analysis of the
structure, identity, binding
affinity and function of the agent. Generally, once identified, the bioactive
agent is resynthesized and
combined with the target cell to verify the exocytosis modulation under
various conditions and in the
presence or absence of other various agents. The bioactive can be prepared in
a therapeutically
effective amount to modulate exocytosis and combined with a suitable
pharmaceutical carrier.

In a preferred embodiment, the cell populations can be subjected to various
experimental conditions,
with and without the candidate agents, and with and without exocytic
stimulation or inhibition.
Changes in conditions include but are not limited to changes in pH,
temperature, buffer or salt
concentration, etc. In a preferred embodiment, the pH is changed, generally by
increasing or
decreasing the pH, usually by from about 0.5 to about 3 pH units.
Alternatively, the temperature is
altered, with increases or decreases of from about 5 C to about 30 C being
preferred. Similarly, the
salt concentration may be modified, with increases or decreases of from about
0.1 M to about 2 M
being preferred.

43


CA 02325597 2008-09-10
77245-54

In a preferred embodiment, the cellular phenotype to be modulated is small
molecule (or other
candidate agent) toxicity. These are generally as outlined above for cell
viability assays. Small
molecule dose responses can also be compared by comparing the cells with the
greatest functional
response, and then backgating to see if there is more or less toxicity
associated with those cells.
In a preferred embodiment, the cellular phenotype involves the expression or
activity of cell surface
receptors; up to sixteen cell surface markers may be followed simultaneously,
with up to eight being
preferred. The presence or absence of any particular cell surface marker can
be detected by directly
and indirectly conjugated antibodies against any cell surface protein whose
cell surface expression
reflects an important functional parameter associated with the cells being
studied. The effect of
candidate agents such as small molecules can then be tested against individual
or multiple markers.
In a preferred embodiment, the cellular phenotype involves the expression or
activity of enzymes such
as fluorescent based reporter systems that can reporta biological event that
occurs simultaneously
with the primary measurement or is a result of the primary measurement. This
reporter system can be
a readout of upstream signal transduction pathways that are active in the
cytoplasm, or of nucleoar
transcriptional or translational events, as well as export events from the
nucleus or the cell.

In a preferred embodiment, the cellular phenotype involves protein-protein
interactions (or interactions
between other binding ligands), such as dimerization, that can be either
disrupted or instigated by a
candidate agent. These events may be measured by the appearance or
disappearance of FRET
between two labeled binding ligands.

The following examples serve to more fully describe the manner of using the
above-described
invention, as well as to set forth the best modes contemplated for carrying
out various aspects of the
invention. It is understood that these examples in no way serve to limit the
true scope of this invention,
but rather are presented for illustrative purposes.

EXAMPLE 1
Cell Cycle Assays using p21 as a positive control
Materials and Methods:
Vector Construction: The coding region of the p21 gene was cloned from Jurkat
cDNA by PCR with an
upstream primer covering the start methionine (5'-GATCGGATCCACC
ACCATGGGCTCAGAACCGGCTGGGGATGTC) and C-terminus (5'-GATCCC
AATTTAATGGTTTTATTTGTCATCGTCATCCTTGTAGTCGGGCTTCCTCTTGGAGAAGATCAGCCG

44


CA 02325597 2000-10-16

WO 99/54494 PCTIUS99/08345
GCGTTTG). The single PCR product was directionally cloned into the CRU5-GFP
retroviral vector
(Rigel, Inc.) through flanking BstXI sites within the primers. The resultant
construct, CRU5-GFP-p21 F
(Figure 1), encodes the GFP fused (in frame) to the human p21 protein with a
Gly insertion at position
2 and a FLAG-epitope at the C-terminus. The C-terminal 24 amino acids of p21
were cloned into the
CRU5-GFP retroviral vector (Rigel, Inc.) through flanking BstXI sites within
the PCR primers: 5'
GATCCCACCACCATGGGCAAACGGCGGCAGACCAGCATGACAGATTTCTACCACTCCAAACGCC
GGCTGATCTTCTCCAA;
5'GATCCCAATTTAAATGGTTTTATTTGTCATCGTCATCCTTGTAGTCGGGCTTCCTCTTGGAGAAG
ATCAGCCGGCGTTTG. The resultant construct, CRU5-GFPp21C (Figure 1), encodes GFP
fused in-
frame to KRRQTSMTDFYHSRRLIFSKRKP and a FLAG-epitope at the C-terminus. The C-
terminal 24
amin acids of p21,, with three alanine mutations, were cloned into the CRU5-
GFP retroviral vector
(Rigel, Inc.) through flanking BstXI sites within the PCT primers:
5'ATCGGATCCACCACCATGGGCAAACGGCGGCAGACCAGCGCCACAGCTGCCTACCACTCC;
5'GATCCCAATTTAATGGTTTTATTTGTCATCGTCATCCTTGTAGTCGGGCTTCCTCTTGGAGAAGA
TCAGCCGGCGTTTG. The resultant construct, CRU5-GFPp21Cmut (Figure 1), encodes
GFP fused
in-frame to KRRQTSATAAYHSRRLIFSKRKP (mutations are underlined) and a FLAG-
epitope at the
C-terminus.

Retroviral Transduction: Phoenix E cells were plated in 6-well plates at 106
cells in 1.5 ml complete-
DMEM (DMEM + 10% FBS + Pen/Strep) and incubated at 37 C for 16 hours. CaCI2-
precipitation
transfection was performed (2 pl DNA (1 pg/pl), 30.5 pl 2M CaCl2, 217.5 pl
H2O, 0.5 ml 2X HBS) with
the CRU5-IRES-GFP vector or CRU5-p2lF-IRES-GFP clone in the presence of 50 pM
chloroquine for
8 hours at 37 C. The transfection-medium was removed and replaced with 2 ml
complete-DMEM and
the cells were further incubated for 16 hours at 37 C. The medium was changed
to 1.5 ml complete-
RPMI (RPMI + 10% FBS + Pen/Strep) and incubated at 32 C for 48 hours. The
virus supernatant
from transfected plates was filtered (0.45pm) and transferred to a 6-well
plate. An 100 NI aliquot
(5x106 cells) of Jurkat T-cells expressing the ecotrophic receptor (JurkatE)
was added to each well.
Polybrene was added to a final concentration of 5 pg/ml. The plates were
sealed with parafilm and
centrifuged at 32 C for 90 minutes at 2500 RPM. The parafilm was removed and
the plate incubated
overnight at 37 C. The medium was changed after 16 hours to 4 ml complete-RPMI
and incubated at
37 C for 72 hours.

Cell Cycle FACS-Assay: The retroviral vector-transduced cells were pelleted
and resuspended at 106
cells/ml in complete-RPMI. One volume (1mi) of 4 pM PKH26 cell tracking dye
(Sigma) was added to
the cells and incubated at 25 C for 5 minutes. The suspension was diluted 5-
fold and the cells
pelleted at 400 x g for 10 minutes at 25 C. The cells were further washed
twice with 6 ml complete-
RPMI and incubated at 3x105 cells/ml in a 6-well plate for 72 hours. The
labeled cells were pelleted


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
and resuspened at 106 cells/ml in complete-RPMI containing 5 ug/ml Hoechst
33342 (Molecular
Probes) and incubated at 37 C for 2 hours. The stained cells were pelleted and
resuspened at >106
cells/ml in FACS buffer (PBS/0.5%FCS/5ug/ml Hoechst 33342). The cells were
subjected to flow-
cytometric analysis on a MoFlo cytometer (Cytomation) equipped with three
lasers. Forward and side
scatter were triggered with a 488nm-line argon laser and scattered light was
collected with a forward
scatter detector and 488nm band pass filter. GFP was excited with a 488nm-fine
argon laser and
emitted light was collected through a 530nm-band pass filter. PKH26 cell
tracking dye was excited
With a 533nm-line HeNe-laser and emitted light was collected through a 570nm-
band pass filter.
Hoechst 33342 dye was excited with a UV-laser and emitted light was collected
through a 450nm-
band pass filter.

Results:
Jurkat T-cells were transduced with with retroviral vectors encoding human p21
(Gp21), or the PCNA
binding C-terminal 24 amino acids (Gp2lC) fused to GFP (Figure 1). A non-PCNA
binding mutant
version of the p21 C-terminal 24 amino acids (Gp2lCmut, Cayrol et al.,
Oncogene 16:311 (1998))
served as a negative control. Expression of the transduced p21 could be
distinguished from the
endogenous protein by the FLAG-epitope by Western blotting (not shown).
Expression of the fusion
proteins was reported in the FACS by GFP fluorescence (Figure 26). Transduced
cells were pulsed
labeled with a cell tracking compound, pkh26, which incorporates red
fluorescent aliphatic molecules
into the cell membrane by selective partioning, allowing a correlation between
cell cycling and
fluorescent intensity: arrested cells remain cell tracker dye bright; cycling
cells dilute the signal and
dim. As shown in Figure 2C, live GFP-p21-expressing cells gated on GFP,
demonstrated a higher red
fluorescence than vector transduced cells expressing identical GFP levels,
indicating cell cycle
arrests. A similar effect was seen in the Gp2lC expressing cells, however,
Gp21-Cmut was identical
to non-expressing cells. The DNA content of the same GFP-gated cells is shown
in Figure 2D. Gp21
expressing cells are arrested in the G1 phase of the cell cycle, Gp21 C-
expressing cells show G1 and
G2 checkpoint accumulation, consistent with previous results (Wade Harper, et
a/., 1993; Cayrol et aL,
1998). The Gp2l Cmut expressing cells show a normal cell cycle distribution.
Viable, arrested,
expressing cells (satisfying the three initial parameters) were sorted based
on DNA content into
separate chambers: left deflection, G1; right deflection, G2.

EXAMPLE 2
Population Based Exocytic Enzyme Activity Measurements

Materials: All chemicals were obtained from Sigma Chemical Co. Dyes and
glucuronide were
obtained from Molecular Probes, Inc. Cell lines MC-9 and RBL-2H3 were obtained
from American
Type Culture Collection (ATCC). Cell culture reagents were obtained from
Fisher Scientific and

46


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
molecular biology reagents from Clontech Inc.

Cell Culture: MC-9 cells were maintained as suspension cultures in flasks in
media consisting of
DMEM with L-arginine (116 mg/ml), L-asparagine (36 mg/ml), sodium pyruvate (1
mM), non-essential
amino acids (0.1 mM), folic acid (6 mg/ml), 2-mercaptoethanol (0.05 mM), L-
glutamine (2 mM), heat
inactivated fetal bovine serum (10%), and 10% T-stim conditioned media
(Collaborative Research,
Inc.). The cells were kept at a density of between .25 and 2 x 106 /ml.
Experiments were only
conducted on cells which were greater than 95% viable as determined by trypan
blue exclusion.
RBL-2H3 cells were maintained as adherent cultures on uncoated (tissue culture
treated) flasks in
media consisting of Eagles MEM with 2mM L-Glutamine and Earl's BSS, 15% heat
inactivated fetal
bovine serum. The cells were passaged (.05% trypsin) so that they were not
confluent for more than
one day.

Exocytosis Stimulation Protocol: Experiments were carried out in modified
tyrodes buffer (MT) which
consisted of NaCI (137 mM), KCI (2.7 mM), CaCI2 (1.8 mM), MgCl2 (1 mM),
Glucose (5.6 mM), Hepes
(20 mM, pH 7.4), and bovine serum albumin (0.1%). MC-9 cells were spun at 400
x g and the media
was aspirated. The cells were then washed with MT, respun/aspirated and taken
up in MT at a
density of 5 x 106 cells /ml. Cells were then treated with either DMSO or
ionophore for 30 minutes (or
the time was varied if a timecourse). The cells were then pelleted with the
supernatant collected for
enzymatic analysis; in some cases, the cells then processed for flow
cytometry. All stimulations were
carried out at 37 C. Stimulations of RBL-2H3 cells were carried out by washing
the adherent cells one
time in MT and then adding warmed MT (1 ml/106 cells) containing the stimulus.
The cells were
incubated at 37 C for 30 minutes and the supernatant was harvested for further
analysis. In some of
the examples, (Examples 4-6), the plate bound cells were stained for annexin
and then removed from
the flask using No-Zyme (Collaborative Research, Inc.) for further processing
for flow cytometry. For
stimulation of RBL-2H3 cells with antigen crosslinking the cells were
incubated overnight with IgE
anti-DNP (Sigma Chemical Co.) in complete media at a concentration of 50
ng/ml. The following day
they were washed one time in MT and stimulated as described above with the
exception that bovine
serum albumin coupled to DNP was used as the stimulus at 100 ng/ml.
Pogulation Based Enzyme Assays: Enzyme assays were carried out on cell
supernatants and pellets
following exocytic stimulation. Cell supernatants were harvested after
stimulation, chilled on ice, and
the post 5000 x g spin supernatant was collected for enzyme activity analysis.
Similarly, cell pellets
were collected/lysed in MT containing 0.1 % triton X-100 and the post 5000 x g
spin supernatant was
collected for enzyme activity analysis. For each analysis 100 pi of lysate or
supernatant was mixed
with 100 pi of reaction buffer (40mM Citrate, pH 4.5) containing 2mM substrate
(4-methylumbelliferyl
Q-D Glucuronide [glucuronidase substrate] or 4-methylumbelliferyl N-acetyl [3-
D glucosaminide

47


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
[hexosaminidase substrate]) in a solid black 96 well plate (Costar, Inc,) and
incubated at 37 C for 15
minutes. The plate was read on a fluorescence plate reader (Wallac, Inc.)
using excitation 380
nm/emission 440 nm filters every 3 minutes for five times to obtain an
enzymatic rate; analyses were
carried out in triplicate.
Flow Cytometry: Cells processed for stimulation and staining were taken up in
MT on ice and filtered
through a 100 pm filter prior to cytometry. Cells were analyzed using a
FACSCAN (Becton Dickinson
Inc., laser line 458 nm) or a Mo-Flo (Cytomation, Inc., laser lines 350nM
broadband (UV), 488nm, and
647nm) Cytometer. Cells were sorted, if desired using the Mo-Flo.
Results: The results are shown in Figure 4. Enzymatic activity in the cell
supernatant was measured
for MC9 (A) and RBL 2H3 (B) cells under various conditions. A) MC-9 cells were
stimulated in the
presence of DMSO (-) or 2 pm lonomycin (+) for 30 minutes. The supernatant was
collected and
analyzed for glucuronidase or hexosaminidase activity. Stimulated release of
granule enzymatic
activity is evident. B) RBL-2H3 cells were sensitized for 16 hours with
varying amounts of IgE
anti-DNP and stimulated to exocytose by exposure to increasing amounts of the
antigen BSA-DNP. A
dose response of both antibody and antigen is evident in the measured
supernatant hexosaminidase
activity.

EXAMPLE 3: Mast Cell Exocytic Light Scatter Changes

The cells were prepared as described in Example 2, and light scatter
properties were determined.
Results: The results are shown in Figure 4. Light scatter changes observed on
the flow cytometer
(side scatter vs. forward scatter) are plotted as bivariate histograms for RBL-
2H3 cells (A, D) and
MC-9 cells (B, C, E, F). Cells were stimulated with the ionophore A23187 (0.5
ug/ml) and observed at
various timepoints [0 minutes (A, C), 5 minutes (E), 10 minutes (D), and 30
minutes (B, F)]. Time
dependent scatter changes are evident in both cell lines with significant
changes occuring during the
first 10 minutes which represents the major bolus of exocytosis in these
cells.
EXAMPLE 4: Styryl Dyes Detect Mast Cell Exoc osis by FACS

S yrI Dye Staining: The cells were prepared as described above. Styryl dyes
(FM1-43 or FM4-64;
Molecular Probes, Inc.) were diluted to a final concentration of 250 nM in MT
and were incorporated
into the stimulation buffer (see Example 2). After the stimulation protocol
the cells were spun down,
aspirated and resuspended in fresh ice cold MT. The cells were then ready for
analysis in the flow
cytometer (see Example 2).

48


CA 02325597 2000-10-16

WO 99/54494 PCTIUS99/08345
Results: The results are shown in Figure 6. MC-9 cells were stimulated (blue =
DMSO, red = 2 pM
ionomycin) in the presence either FM 4-64 (A, B) or FM 1-43 (C,D,E). A) FM 4-
64 labeled cells
detected in the flow cytometer in fluorescence channel 1. B) FM 4-64 labeled
cells detected in the flow
cytometer in fluorescence channel 3. C) FM 1-43 labeled cells detected in the
flow cytometer in
fluorescence channel 1. D) FM 1-43 labeled cells detected in the flow
cytometer in fluorescence
channel 3. There is a clear stimulation dependent increase of fluoreceence
intensity with both dyes;
FM 4-64 being the most red-shifted and predominantly detected in channel 3
while FM 1-43 is more
broadly fluorescent being detected in both channels 1 and 3. E) MC-9 cells
were preincubated with
varying doses of the PI-3 kinase inhibitor wortmannin (1 pM-bar/, 100 nM-bar2,
10 nM-bar3, and 0
nM-bars 1 and 2) prior to stimulation with A23187 (0.5ug/ml, bars 1-4) or DMSO
(bar 5) in the
presence of FM 1-43. The mean channel shift detected in the flow cytometer in
fluorescence channel
1 is plotted as a bar graph. Wortmannin, a known inhibitor of mast cell
exocytosis, causes a dose
dependent decrease in the FM 1-43 signal indicating that FM 1-43 signal
reflects the degree of
degranulation in the MC-9 mast cell line.
EXAMPLE 5: Annexin-V Staining Detects Mast Cell Exocytosis by FACS
Materials: Annexin-V biotin, Annexin-V FITC and streptavidin APC were obtained
from Caltag
Laboratories. Other materials and methods used herein can be incorporated from
the other examples,
particularly Example 2.

Annexin-V Staining: Cells post exocytic stimulus were stained with annexin-
PITC at a dilution of 1/100
in MT for 10 minutes at room temperature. The cells were then washed one time
in MT, taken up in
MT and viewed in the flow cytometer or microscope. For indirect labeling,
annexin-biotin was added to
the MT during the stimulation procedure at a dilution of 1/200. The cells were
then pelleted in ice cold
MT, spun, aspirated, and taken up in ice cold MT with streptavidin-APC at a
dilution of 1/200 and kept
on ice for 15 minutes. After pelleting the cells and aspirating the
Streptavidin-APC, the cells were
resuspended in MT and viewed in the flow cytometer. In some experiments
different secondaries
were applied such as streptavidin alexa 488 or 594 (Molecular Probes, Inc.)
for visualization in the
microscope.

Results: The results are shown in Figure 7. MC-9 cells were stimulated with
either DMSO (Figures A
and B) or 2 pm ionomycin (Figures C and D) and then stained with both
propidium iodide [PIj (Figures
A and C) and annexin-V-FITC (Figures B and D). Stimulation with this dose of
ionomycin does not
compromise the plasma membrane as demonstrated by no significant increase in
PI staining in the
exocytosing cells. Degranulation results in a significant increase in annexin
binding as seen
comparing Figures D and B.

49


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
EXAMPLE 6: Annexin-V-FITC stains exocytic granules in MC-9 cells visualized by
confocal
microscopy

Microscopy: Cells post stimulation or staining were mounted onto glass slides
and coverslipped; these
were directly visualized by brighffield and fluorescence microscopy on an
inverted microscope (TE300,
Nikon) using standard BFP, FITC, or TRITC filter sets. Some images were
obtainad using an inverted
confocal scanning microscope (Zeiss, Inc,, Bio-Rad, Inc.) using standard FITC
and TRITC filter sets.

Results: MC-9 cells were stimulated with 2 pm ionomycin or DMSOand stained
with annexin-V-FITC
and mounted for confocal microscopy (data not shown). All viable unstimulated
cells show low
annexin binding and no cell surface granular staining.

EXAMPLE 7: Annexin-V-FITC Stains Exocytic Granules in RBL-2H3 Cells Stimulated
with
Antigen Crosslinking

Except as othewise stated below, the methods for this example are described in
the preceding
examples.

Results: RBL-2H3 cells were sensitized with IgE and stimulated to exocytosa
with either MT buffer
only or BSA-DNP antigen (100 ng/ml) and then stained with annexin-V-FITC.
Numerous cell surface
granules are stained in the antigen stimulated cells similar to the pattern
seen in MC 9-cells. Viable
unstimulated cells show negligible annexin-V staining (data not shown).

EXAMPLE 8: In Situ Enzymology of Exocytosing Cells Visualized in the FACS
in Situ Enzvmology: MC-9 cells were stimulated for exocytosis as described
above and then
incubated in enyme substrate buffer (BSA free MT, pH 4.3 - 7.4 range)
containing the substrate
ELF-97 Glucuronide (250 pM) for 15 minutes at 37 C. The cells were washed one
time in MT and
then viewed in the microscope or on the flowcytometer. Further methodologies
are described in the
preceding examples.

Results: The results are shown in Figure 8. MC-9 cells ware stimulated with
DMSO (Figure A) or
A23187 (0.5 pg/ml - Figure B and C) and then stained for in situ glucuronidase
activty. A) Flow
cytometer histogram of ELF-97 detection indicative of cell surface enzymatic
precipitate. Very low
signal is seen in the DMSO treated cells. B) Flow cytometer histogram of ELF-
97 detection indicative
of cell surface enzymatic precipitate. A significant increase in signal is
seen upon secretory



CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
stimulation with ionophore. C) pH profile of the cell surface enzymatic
activity. MT buffer, prepared at
different pHs, was used to pH profile the signal seen in the flow cytometer.
The bar graphs represent
the percentage of maximal signal (as measured by mean channel shift in the
flow cytomoter)
observed. The enzymatic activity is pH dependent with a peak at less than pH
6; this is consistent with
enzymatic acitivity derived from an acidic secretory granule.

EXAMPLE 9: Lysotracker Green Is Released from Mast Cell Granules upon
Exocytosis and
Can Be Detected by FACS

Lvsotracker Dye Staining: Lysotracker dyes (blue, green, and red) were loaded
into cells by diluting
them to a final concentration of 1 pM in complete media and incubating the
cells for 60 minutes at
37 C in their presence. After loading, the cells were washed two times in MT
and then were ready for
further analysis or stimulation. Further methodologies are described in the
preceding examples.

Results: The results are shown in Figure 9. MC-9 cells were loaded with
Lysotracker green for 1 hour
and then stimulated with either DMSO or ionomycin (2 NM) and viewed in the
flow cytometer. Shown
is a histogram of fluorescence intensity detected in channel 1; a significant
loss of signal is seen in the
ionophore stimulated sample as compared to the DMSO control which is
reflective of the release of
lysotracker green dye from the secretory granules.
EXAMPLE 10: Multiparameter Analysis--Lysotracker Green, Annexin-V-APC,
Forward and Side Scatter

Except as otherwise described below, the methodologies described in the
preceding examples were
used.

Results: The results are shown in Figure 10. MC-9 cells were treated with
different doses of
ionomycin (0 NM- A, E; 1 NM- B, F; 2 NM- C, G; and 3 NM- D, H) and observed in
the flow cytometer
with four parameters simultaneously. The cells were loaded with lysotracker
green for one hour and
then stimulated and stained for annexin-VAPC. Figures A-D: Bivariate
histograms of side vs. forward
light scatter. Note the dose dependent changes in both parameters from left to
right as forward scatter
increases and side scatter decreases. Figures E-H: Bivariate histograms of
annexin-V-APC vs.
Lysotracker green signals. As exocytosis increases (left to right) annexin
signal becomes greater as
the lysotracker signal decreases. This reflects the binding of annexin-V to
the cell surface granules
and the loss of lysotracker from these granules as they are exposed to the
extracellular milieu.
EXAMPLE 11: Multiparameter Analysis--FM 1-43 Annexin-V-APC. Forward and Side
Scatter
51


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
Except as otherwise noted below, the methodologies described above were used
herein.

Results: MC-9 cells were treated with either DMSO or ionomycin (2 NM) and
observed in the flow
cytometer with four parameters simultaneously. The cells were stimulated in
the presence of FM 1-43
and stained for annexin-V-APC (data not shown). There were stimulation
dependent changes in both
parameters at the 30 minute timepoint. There were stimulation dependent
increases in both signals.
The changes reflect the binding of annexin-V to the cell surface granules and
the simultaneous
coupled endocytosis of the FM 1-43 dye into the MC 9 cells.
EXAMPLE 12: Simultaneous Multiparameter Measurements in the FACS Correlate
with
Population Based Enzyme Readouts

Calcium Signaling Assays: MC-9 or RBL-2H3 cells were washed one time in MT and
loaded with the
Ca++ sensitive probe Fluo-3 (1 NM, Molecular Probes, Inc.) in MT at 37 C for
20 minutes. The cells
were washed one time in warm MT and then stimulated using the protocol
described above. The
signal due to rise in the intracellular Ca++ concentration was visualized
using either the flow cytometer
(see below), fluorescence microscopy, or read on a fluorescence plate reader
(Wallac, Inc.). Loading
of the cells was determined by releasing the intracellular dye with MT
containing 0.1 % triton X-1 00.
Except as otherwise noted below, the methodologies described above were used
herein.

Results: The results are shown in Figure 11. MC-9 cells were stimulated in the
presence of FM 1-43
and annexin-V-APC stained as described in the methods above. At various
timepoints after ionomycin
stimulation the cells were put on ice and either analyzed by flow cytometry or
for enzymatic activity
(cell supernatant). The parameters forward scatter, FM 1-43, annexin-V-APC,
and hexosaminidase
are plotted on the graph relative to the maximal response for each parameter.
For calcium signaling, a
separate tube of cells was loaded with Fluo-3 and underwent the identical
procedure. The
timecourses of the cytometry based parameters indicate that they correlate
quite well with exocytosis
as measured by hexosaminidase release. Forward scatter, in this example, shows
an effect which
varies both positively and negatively with time.

EXAMPLE 12: Expression of VAMP-GFP and VAMP-FRET Constructs
cDNA Constructs:
VAMP-GFP construct: The rat VAMP-2 cDNA (obtained from R. Scheller, Stanford
University) was
PCR modified to introduce: (1) a 5' BstXl site encoding a concensus Kozak and
glycine insertion

52


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
(a.a.2) to facilitate expression and in vivo stability, respectively; (2) a
serine-glycine linker with a
BamHl site at the 3' end. The GFP coding sequence from CdimGFP (Clontech,
Inc.) was PCR
modified to introduce a 3' BstXI site encoding a stop codon. The VAMP-GFP
fusion was constructed
by ligating the modified rVAMP and GFP PCR fragments through a common BamHl
site in the
serine-glycine linker to create an in-frame fusion protein with the following
sequence:
MGSATAATVPPAAPAGEGGPPAPPPNLTSNRRLQQTQAQVDEWDIMRVNVDKVLERDQKLSELDD
RADALQAGASQFETSAAKLKRKYWWKNLKMMI I LGVICAII Lill IVYFST
GSGSGSGSGSGPVSKGEELFTGWPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTTGKLPV
PWPTLVTTLTHGVQCFSRYPDHMKQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVN
RIELKGIDFKEDGNILGHKLEYNFNSHNVYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGD
GPVLLPDNHYLSTQSALSKDPNEKRDHMVLLEFVTAAGITLGMDELYKZ
The VAMP sequence is underlined, the serine-glycine linker is italicized and
the GFP sequence is in
regular text.

The VAMP-GFP fusion sequence was cloned into the 96.7 retrovlral vector with
directional BstXI sites
to create pVG. The sequence was verified by sequencing in both directions.
Proper expression was
verified in transfected and infected cells by Western analysis and
fluorescence microscopy.
Trp-FRET construct: The GFP coding sequence from cGFP (Clontech, Inc.) was PCR
modified to
create: (1) a 5' BstXI site encoding a concensus Kozak and glycine insertion
(a.a.2) to faciltate
expression and in vivo stability, respectively; (2) a 3'-end Sacli site
encoding A1a228 at the C-terminus.
The BFP coding sequence from cBFP (Clontech, Inc.) was PCR modified to create:
(1) a 5' BamHl
site encoding Ser2; (2) a 3'-end BstXI encoding a stop codon. A SaclI-BamHl
conversion linker
encoding Factor X and tryptase protease cleavage sites, flanked by GSGS
spacers
(GSGSIEGRLRKQGSCS) was used to fuse the GFP and BFP to create an in-frame
fusion protein with
the following sequence:

IV KGEELFTGWPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTLTYG
VQCFSRYPDHMKQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDG
NILGHKLEYNYNSHNVYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHYLST
QSALSKDPNEKRDHMVLLEFVTAAGSGS/EGRLRKQGSGSKGEELFTGWPILVELDGDVNGHKFSV
SGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTLTHGVQCFSRYPDHMKQHDFFKSAMPEGYVQER
TI FFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNFNSHNVYIMADKQKNGIKVNFK
IRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHYLSTQSALSKDPNEKRDHMVLLEFVTAAGITLGMD
ELYKZ

53


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
The GFP sequence is underlined, the Factor X/tryptase site linker is
italicized and the BEP sequence
is in regular text.

The VAMP-GFP fusion sequence was cloned into the BamHl and BstXl sites of the
retroviral vector
96.7 to create pGX/TB. The sequence was verified by sequencing in both
directions. Proper
expression was verified in transfected and infected cells by Western analysis
and fluorescence
microscopy.

The VAMP-GFP encoding sequence was PCR modified to create a 3'-end Sacll site
encoding A1a228
at the C-terminus. This fragment was cleaved with Xhol and Sacll and cloned
into the Xhol/Sacll sites
of pGX/TB to create pVGX/TB (Trp-FRET), encoding the rVAMP-2-BFP-
FactorX/Tryptase sites-GFP
fusion protein with the following sequence:
GSATAATVPPAAPAGEGGPPAPPPNLTSNRRLQQTQAQVDEWDIMRVNVDKVLERDQKLSELDDR
ADALQAGASQFETSAAKLKRKYVWVKN LKMMI ILGVICAI I LI I I IVYFSTGSGSGSGSGSGPV
SKGEELFTGWPILVELDGDVNGHKFSVSGEGEGDATYGKLTLKFICTTGKLPVPWPTLVTTLTHGVQ
CFSRYPDHMKQHDFFKSAMPEGYVQERTIFFKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNIL
GHKLEYNFNSHNVYIMADKQKNGIKVNFKIRHNIEDGSVQLADHYQQNTPIGDGPVLLPDNHYLSTQS
ALSKDPNEKRDHMVLLEFVTAAGSGSIEGRRKLQGSGSKGEELFTGWPILVELDGDVNGHKFSVSG
EGEGDATYGKLTLKI FCTTGKLPVPWPTLVTTLTYGVQCFSRYPDHMKQHDFFKSAMPEGYVQERTI F
FKDDGNYKTRAEVKFEGDTLVNRIELKGIDFKEDGNILGHKLEYNYNSHNVYIMADKQKNGIKVNFKIR
HNIEDGSVQLADHYQQNTPIGDGPVLLPDNHYLSTQSALSKDPNEKRDHMVLLEFVTAAGITLGMDEL
YKZ

The VAMP sequence is underlined, the serine-glycine linker is italicized, the
Factor X/tryptase site
linker is in bold and the GFP and BFP sequences are in shown in regular text.

The rVAMP-2-BFP-FactorX/Tryptase sites-GFP fusion sequence was verified by
sequencing in both
directions. Proper expression was verified in transfected and infected cells
by Western and
fluorescence microscopy and FACS analysis.
Transfections and Infections: To infect MC-9 and RBL-2H3 cells with
recombinant retroviruses
expressing the Vamp constructs the following procedure was carried out Phoenix
E or A cells
(obtained from G. Nolan, Stanford Univ.) were plated out in 6 well plates at 8
x 10E5 cells in 1.5 ml
media (DMEM, 10% FBS) on day one. On day two 5pg of DNA was transfected into
the cells using
the CaPO4 precipitation method in the presence of 50 pM chioroquine. The
precipitate was incubated
with the cells for 8 hours at 37 C at which time the medium was removed,
washed once with fresh
media and replaced with either fresh MC-9 or RBL-2H3 media; the cells were
then incubated at 32 C

54


CA 02325597 2000-10-16

WO 99/54494 PCT/US99/08345
for 48-72 hours. The supernatant from the Phoenix cells (viral supematant) was
spun at 1000 xg for
minutes and protamine sulfate was added to a final concentration of 5 pg/ml;
this supernatant was
added to the MC-9 or RBL-2HS freshly trypsinized) cells in a 6 well plate (5 x
10E5 cells per well) and
the mixture was spun at 1000 x g for 90 minutes at room temperature The cells
were then incubated at
5 32 C for 16 hours. The viral supernatant was removed and fresh media was
added; target gene
expression was seen after 24 hours post infection.



CA 02325597 2001-03-21
SEQUENCE LISTING
<110> Rigel Pharmaceuticals, Inc.

<120> Multiparameter Facs Assays to Detect Alterations in
Cellular Parameters and to Screen Small Molecule
Libraries

<130> FP68104-PC/DJB/RMS
<140> PCT/US99/08345
<141> 1999-04-16

<160> 55

<170> Patentln Ver. 2.0
<210> 1
<211> 27
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 1
Lys Arg Arg Gln Thr Ser Met Thr Ser Met Thr Asp Phe Tyr His Ser
1 5 10 15
Lys Arg Arg Leu Ile Phe Ser Lys Arg Lys Pro
20 25
<210> 2
<211> 27
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 2
Lys Arg Arg Gln Thr Ser Ala Thr Ser Met Ala Ala Phe Tyr His Ser
1 5 10 15
Lys Arg Arg Leu Ile Phe Ser Lys Arg Lys Pro
20 25

<210> 3
<211> 9
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 3
Arg Thr Val Leu Gly Val Ile Gly Asp
1 5

1


CA 02325597 2001-03-21
<210> 4
<211> 9
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 4
Arg Thr Ala Leu Gly Asp Ile Gly Asn
1 5
<210> 5
<211> 27
<212> PRT
<213> Rat
<400> 5
Tyr Met Thr Val Ser Ile Ile Asp Arg Phe Met Gln Asp Ser Cys Val
1 5 10 15
Pro Lys Lys Met Leu Gln Leu Val Gly Val Thr
20 25
<210> 6
<211> 28
<212> PRT
<213> Rat

<400> 6
Lys Phe Arg Leu Leu Gln Glu Thr Met Tyr Met Thr Val Ser Ile Ile
1 5 10 15
Asp Arg Phe Met Gln Asn Ser Cys Val Pro Lys Lys
20 25
<210> 7
<211> 27
<212> PRT
<213> Rat
<400> 7
Arg Ala Ile Leu Ile Asp Trp Leu Ile Gln Val Gln Met Lys Phe Arg
1 5 10 15
Leu Leu Gln Glu Thr Met Tyr Met Thr Val Ser
20 25
<210> 8
<211> 27
<212> PRT
<213> Rat
<400> 8
Asp Arg Phe Leu Gln Ala Gln Leu Val Cys Arg Lys Lys Leu Gln Val
1 5 10 15
Val Gly Ile Thr Ala Leu Leu Leu Ala Ser Lys
20 25

2


CA 02325597 2001-03-21
<210> 9
<211> 18
<212> PRT
<213> Rat
<400> 9
Met Ser Val Leu Arg Gly Lys Leu Gln Leu Val Gly Thr Ala Ala Met
1 5 10 15
Leu Leu

<210> 10
<211> 61
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> EMBO J.
<304> 13
<305> 22
<306> 5303-5309
<307> 1994
<400> 10
Met Gly Cys Ala Ala Leu Glu Ser Glu Val Ser Ala Leu Glu Ser Glu
1 5 10 15
Val Ala Ser Leu Glu Ser Glu Val Ala Ala Leu Gly Arg Gly Asp Met
20 25 30
Pro Leu Ala Ala Val Lys Ser Lys Leu Ser Ala Val Lys Ser Lys Leu
40 45

Ala Ser Val Lys Ser Lys Leu Ala Ala Cys Gly Pro Pro
50 55 60

<210> 11
<211> 6
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 11
Gly Arg Gly Asp Met Pro
1 5
<210> 12
<211> 69
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
3


CA 02325597 2001-03-21
<400> 12
Met Gly Arg Asn Ser Gln Ala Thr Ser Phe Gly Thr Phe Ser His Phe
1 5 10 15
Tyr Met Glu Trp Val Arg Gly Gly Glu Tyr Ile Ala Ala Ser Arg His
20 25 30
Lys His Asn Lys Tyr Thr Thr Glu Tyr Ser Ala Ser Val Lys Gly Arg
35 40 45
Tyr Ile Val Ser Arg Asp Thr Ser Gln Ser Ile Leu Tyr Leu Gln Lys
50 55 60
Lys Lys Gly Pro Pro
<210> 13
<211> 7
20 <212> PRT
<213> Monkey virus
<300>
<301> Kalderon et al.,
<303> Cell
<304> 39
<306> 499-509
<307> 1984

30 <400> 13
Pro Lys Lys Lys Arg Lys Val
1 5
<210> 14
<211> 6
<212> PRT
<213> Homo sapiens
40 <400> 14
Ala Arg Arg Arg Arg Pro
1 5
<210> 15
<211> 10
<212> PRT
<213> Artificial Sequence
50 <220>
<223> Description of Artificial Sequence: synthetic
<300>
<301> Ghosh et al.,
<303> Cell
<304> 62
<306> 1019-
<307> 1990

60 <400> 15
Glu Glu Val Gln Arg Lys Arg Gln Lys Leu
1 5 10
4


CA 02325597 2001-03-21
<210> 16
<211> 9
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence; synthetic
<300>
<301> Nolan et al.,
<303> Cell
<304> 64
<305> 961
<307> 1991
<400> 16
Glu Glu Lys Arg Lys Arg Thr Tyr Glu
1 5
<210> 17
<211> 20
<212> PRT
<213> African clawed toad
<300>
<301> Dingwell et al.,
<303> Cell
<304> 30
<306> 449-458
<307> 1982
<300>
<301> Dingwell et al.,
<303> J. Cell Biol.
<304> 107
<306> 641-849
<307> 1988
<400> 17
Ala Val Lys Arg Pro Ala Ala Thr Lys Lys Ala Gly Gln Ala Lys Lys
1 5 10 15
Lys Lys Leu Asp
<210> 18
<211> 31
50 <212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<301> Nakauchi et al.,
<303> Proc. Natl. Acad. Sci. U.S.A.
<304> 82
60 <306> 5126-
<307> 1985

5


CA 02325597 2001-03-21
<400> 18
Met Ala Ser Pro Leu Thr Arg Phe Leu Ser Leu Asn Leu Leu Leu Leu
1 5 10 15
Gly Glu Ser Ile Leu Gly Ser Gly Glu Ala Lys Pro Gln Ala Pro
20 25 30
<210> 19
<211> 22
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<301> Staunton et al.,
<303> Nature
<304> 339
<306> 61-
<307> 1989
<400> 19
Met Ser Ser Phe Gly Tyr Arg Thr Leu Thr Val Ala Leu Phe Thr Ile
1 5 10 15
Leu Ile Cys Cys Pro Gly
30

<210> 20
<211> 51
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
40 <300>
<301> Nakauchi et al.,
<303> Proc. Natl. Acad. Sci. U.S.A.
<304> 82
<306> 5126-
<307> 1985
<400> 20
Pro Gln Arg Pro Glu Asp Cys Arg Pro Arg Gly Ser Val Lys Gly Thr
1 5 10 15
Gly Leu Asp Phe Ala Cys Asp Ile Tyr Ile Trp Ala Pro Leu Ala Gly
20 25 30
Ile Cys Val Ala Leu Leu Leu Ser Leu Ile Ile Thr Leu Ile Cys Tyr
35 40 45
His Ser Arg
60
<210> 21
<211> 33

6


CA 02325597 2001-03-21
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<301> Staunton et al.,
<303> Nature
<304> 339
<306> 61-
<307> 1989
<400> 21
Met Val Ile Ile Val Thr Val Val Ser Val Leu Leu Ser Leu Phe Val
1 5 10 15
Thr Ser Val Leu Leu Cys Phe Ile Phe Gly Gln His Leu Arg Gln Gln
25 30
Arg
<210> 22
<211> 37
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<301> Homans et al.,
<303> Nature
<304> 333
<305> 6170
<306> 269-272
<307> 1988
<300>
<303> J. Biol. Chem.
<304> 266
<306> 1250-
<307> 1991
<400> 22
Pro Asn Lys Gly Ser Gly Thr Thr Ser Gly Thr Thr Arg Leu Leu Ser
1 5 10 15
Gly His Thr Cys Phe Thr Leu Thr Gly Leu Leu Gly Thr Leu Val Thr
20 25 30
Met Gly Leu Leu Thr
<210> 23
<211> 14
<212> PRT
60 <213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
7


CA 02325597 2001-03-21
<300>
<303> Mol. Cell. Biol.
<304> 4
<305> 9
<306> 1834-
<307> 1984
<300>
<303> Science
<304> 262
<306> 1019-1024
<307> 1993
<400> 23
Met Gly Ser Ser Lys Ser Lys Pro Lys Asp Pro Ser Gln Arg
1 5 10
<210> 24
<211> 26
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> J. Biol. Chem.
<304> 269
<306> 27791-
<307> 1994
<400> 24
Leu Leu Gln Arg Leu Phe Ser Arg Gln Asp Cys Cys Gly Asn Cys Ser
1 5 10 15
Asp Ser Glu Glu Glu Leu Pro Thr Arg Leu
20 25
<210> 25
<211> 20
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> J. Mol. Neurosci.
<304> 5
<305> 3
<306> 207-
<307> 1994
<400> 25
Lys Gln Phe Arg Asn Cys Met Leu Thr Ser Leu Cys Cys Gly Lys Asn
1 5 10 15
Pro Leu Gly Asp

8


CA 02325597 2001-03-21
<210> 26
<211> 19
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Nature
<304> 302
<306> 33-
<307> 1983
<400> 26
Leu Asn Pro Pro Asp Glu Ser Gly Pro Gly Cys Met Ser Cys Lys Cys
1 5 10 15
Val Leu Ser

<210> 27
<211> 5
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Ann. N. Y. Acad. Sci.
<304> 674
<306> 58-
<307> 1992
<400> 27
Lys Phe Glu Arg Gln
1 5
<210> 28
<211> 36
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Cell. Mol. Biol. Res.
<304> 41
<306> 405-
<307> 1995
<400> 28
Met Leu Ile Pro Ile Ala Gly Phe Phe Ala Leu Ala Gly Leu Val Leu
1 5 10 15
Ile Val Leu Ile Ala Tyr Leu Ile Gly Arg Lys Arg Ser His Ala Gly
20 25 30
Tyr Gln Thr Ile

9


CA 02325597 2001-03-21
<210> 29
<211> 35
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Biochem. Biophys. Res. Commun.
<304> 205
<306> 1-5
<307> 1994
<400> 29
Leu Val Pro Ile Ala Val Gly Ala Ala Leu Ala Gly Val Leu Ile Leu
1 5 10 15
Val Leu Leu Ala Tyr Phe Ile Gly Leu Lys His His His Ala Gly Tyr
20 25 30
Glu Gln Phe
<210> 30
<211> 27
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Eur. J. Biochem.
<304> 165
<306> 1-6
<307> 1987
<400> 30
Met Leu Arg Thr Ser Ser Leu Phe Thr Arg Arg Val Gln Pro Ser Leu
1 5 10 15
Phe Ser Arg Asn Ile Leu Arg Leu Gln Ser Thr
20 25
<210> 31
<211> 25
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Eur. J. Biochem.
<304> 165
<306> 1-6
<307> 1987



CA 02325597 2001-03-21
<400> 31
Met Leu Ser Leu Arg Gln Ser Ile Arg Phe Phe Lys Pro Ala Thr Arg
1 5 10 15
Thr Leu Cys Ser Ser Arg Tyr Leu Leu
20 25
<210> 32
<211> 64
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Eur. J. Biochem.
<304> 165
<306> 1-6
<307> 1987
<400> 32
Met Phe Ser Met Leu Ser Lys Arg Trp Ala Gln Arg Thr Leu Ser Lys
1 5 10 15
Ser Phe Tyr Ser Thr Ala Thr Gly Ala Ala Ser Lys Ser Gly Lys Leu
20 25 30
Thr Gln Lys Leu Val Thr Ala Gly Val Ala Ala Ala Gly Ile Thr Ala
40 45

Ser Thr Leu Leu Tyr Ala Asp Ser Leu Thr Ala Glu Ala Met Thr Ala
50 55 60
<210> 33
<211> 41
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Eur. J. Biochem.
<304> 165
<306> 1-6
<307> 1987
<400> 33
Met Lys Ser Phe Ile Thr Arg Asn Lys Thr Ala Ile Leu Ala Thr Val
1 5 10 15
Ala Ala Thr Gly Thr Ala Ile Gly Ala Tyr Tyr Tyr Tyr Asn Gln Leu
20 25 30
Gln Gln Gln Gln Gln Arg Gly Lys Lys
35 40

<210> 34
<211> 4

11


CA 02325597 2001-03-21
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Royal Society London Transaction B
<306> 1-10
<307> 19992
<400> 34
Lys Asp Glu Leu
1

<210> 35
<211> 15
<212> PRT
<213> adenovirus
<300>
<303> EMBO J.
<304> 9
<306> 3153-
<307> 1990
<400> 35
Leu Tyr Leu Ser Arg Arg Ser Phe Ile Asp Glu Lys Lys Met Pro
1 5 10 15
<210> 36
<211> 19
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Nature
<304> 302
<306> 33-
<307> 1983
<400> 36
Leu Asn Pro Pro Asp Glu Ser Gly Pro Gly Cys Met Ser Cys Lys Cys
1 5 10 15
Val Leu Ser
<210> 37
<211> 15
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Proc. Natl. Acad. Sci. U.S.A.

12


CA 02325597 2001-03-21
<304> 91
<306> 11963-
<307> 1994
<400> 37
Leu Thr Glu Pro Thr Gln Pro Thr Arg Asn Gln Cys Cys Ser Asn
1 5 10 15
<210> 38
<211> 9
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> EMBO J.
<304> 1
<306> 13053-
<307> 1996
<400> 38
Arg Thr Ala Leu Gly Asp Ile Gly Asn
1 5
<210> 39
<211> 29
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Nucleic Acids Res.
<304> 7
<306> 30-
<307> 1979
<400> 39
Met Ala Thr Gly Ser Arg Thr Ser Leu Leu Leu Ala Phe Gly Leu Leu
1 5 10 15
Cys Leu Pro Gln Leu Gln Glu Gly Ser Ala Phe Pro Thr
20 25
<210> 40
<211> 27
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Nature
<304> 284
<306> 26-
<307> 1980

13


CA 02325597 2001-03-21
<400> 40
Met Ala Leu Trp Met Arg Leu Leu Pro Leu Leu Ala Leu Leu Ala Leu
1 5 10 15
Trp Gly Pro Asp Pro Ala Ala Ala Phe Val Asn
20 25
<210> 41
<211> 18
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<300>
<303> Proc. Natl. Acad. Sci. U.S.A.
<304> 80
<306> 3563-
<400> 41
Met Lys Ala Lys Leu Leu Val Leu Leu Tyr Ala Phe Val Ala Gly Asp
1 5 10 15
Gln Ile

<210> 42
<211> 24
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 42
Met Gly Leu Thr Ser Gln Leu Leu Pro Pro Leu Phe Phe Leu Leu Ala
1 5 10 15
Cys Ala Gly Asn Phe Val His Gly

<210> 43
<211> 10
<212> PRT
<213> Artificial Sequence
50 <220>
<221> UNSURE
<222> (3)..(6)
<223> The x at position 3, 4, 5 and 6 represents any
amino acid.

<220>
<223> Description of Artificial Sequence: synthetic
<400> 43
60 Met Gly Xaa Xaa Xaa Xaa Gly Gly Pro Pro
1 5 10
14


CA 02325597 2001-03-21
<210> 44
<211> 5
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 44
Gly Ser Gly Gly Ser
1 5
<210> 45
<211> 4
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 45
Gly Gly Gly Ser
1

<210> 46
<211> 43
<212> DNA
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 46
gatcggatcc accaccatgg gctcagaacc ggctggggat gtc 43
<210> 47
<211> 79
<212> DNA
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 47
gatcccaatt taatggtttt atttgtcatc gtcatccttg tagtcgggct tcctcttgga 60
gaagatcagc cggcgtttg 79
<210> 48
<211> 81
<212> DNA
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 48
gatcccacca ccatgggcaa acggcggcag accagcatga cagatttcta ccatctccaa 60
acgccggctg atcttctcca a 81


CA 02325597 2001-03-21
<210> 49
<211> 80
<212> DNA
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 49
gatcccaatt taaatggttt tatttgtcat cgtcatcctt gtagtcgggc ttcctcttgg 60
agaagatcag ccggcgtttg 80
<210> 50
<211> 60
<212> DNA
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 50
atcggatcca ccaccatggg caaacggcgg cagaccagcg ccacagctgc ctaccactcc 60
<210> 51
<211> 79
<212> DNA
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 51
gatcccaatt taatggtttt atttgtcatc gtcatccttg tagtcgggct tcctcttgga 60
gaagatcagc cggcgtttg 79
<210> 52
<211> 365
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 52
Met Gly Ser Ala Thr Ala Ala Thr Val Pro Pro Ala Ala Pro Ala Gly
1 5 10 15
Glu Gly Gly Pro Pro Ala Pro Pro Pro Asn Leu Thr Ser Asn Arg Arg
20 25 30
Leu Gln Gln Thr Gln Ala Gln Val Asp Glu Val Val Asp Ile Met Arg
35 40 45
Val Asn Val Asp Lys Val Leu Glu Arg Asp Gln Leu Ser Glu Leu Asp
50 55 60

Asp Arg Ala Asp Ala Leu Gln Ala Gly Ala Ser Gln Phe Glu Thr Ser
70 75 80
16


CA 02325597 2001-03-21

Ala Ala Lys Leu Lys Arg Lys Tyr Trp Trp Lys Asn Leu Met Met Ile
85 90 95
Ile Leu Gly Val Ile Cys Ala Ile Ile Leu Ile Ile Ile Ile Val Tyr
100 105 110
Phe Ser Thr Gly Ser Gly Ser Gly Ser Gly Ser Gly Ser Gly Pro Val
115 120 125

Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu Val Glu
130 135 140
Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly Glu Gly
145 150 155 160
Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile Cys Thr
165 170 175
Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr Leu Thr
180 185 190

His Gly Val Gln Cys Phe Ser Arg Tyr Pro Asp His Met Lys Gln His
195 200 205
Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu Arg Thr
210 215 220
Ile Phe Phe Lys Asp Asp Gly Asn Tyr Thr Arg Ala Glu Val Lys Phe
225 230 235 240
Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly Ile Asp Phe
245 250 255
Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr Asn Phe Asn
260 265 270
Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn Gly Ile Lys
275 280 285

Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser Val Gln Leu
290 295 300
Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly Pro Val Leu
305 310 315 320
Leu Pro Asp Asn His Tyr Leu Ser Thr Gln Ser Ala Leu Ser Lys Asp
325 330 335
Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe Val Thr Ala
340 345 350

Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys Glx
355 360 365
<210> 53
<211> 16
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
17


CA 02325597 2001-03-21
<400> 53
Gly Ser Gly Ser Ile Glu Gly Arg Leu Arg Lys Gln Gly Ser Cys Ser
1 5 10 15
<210> 54
<211> 477
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 54
Met Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu
1 5 10 15
Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly
25 30
Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile
35 40 45
Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr
50 55 60
Leu Thr Tyr Gly Val Gln Cys Phe Ser Arg Tyr Pro Asp His Met Lys
65 70 75 80

Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu
85 90 95
Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu
100 105 110
Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Gly Ile
115 120 125

Asp Phe Lys Glu Asp Gly Met Ile Leu Gly His Lys Leu Glu Tyr Asn
130 135 140

Tyr Asn Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn Gly
145 150 155 160
Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser Val
165 170 175

Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly Pro
180 185 190
Val Leu Leu Pro Asp Asn His Tyr Leu Ser Thr Gln Ser Ala Leu Ser
195 200 205
Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe Val
210 215 220
Thr Ala Ala Gly Ser Gly Ser Ile Glu Gly Arg Leu Arg Lys Gln Gly
225 230 235 240

Ser Gly Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu
245 250 255
18


CA 02325597 2001-03-21

Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly
260 265 270
Glu Gly Glu Gly Asp Ala Thr Gly Lys Leu Thr Leu Lys Phe Ile Cys
275 280 285
Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr Leu
290 295 300

Thr His Gly Val Gln Cys Phe Ser Arg Tyr Pro Asp His Met Lys Gln
305 310 315 320
His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu Arg
325 330 335

Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu Val
340 345 350
Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly Ile
355 360 365
Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr Asn
370 375 380

Phe Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn Gly Ile
385 390 395 400
Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser Val Gln
405 410 415
Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly Pro Val
420 425 430
Leu Leu Pro Asp Asn His Tyr Leu Ser Thr Gln Ser Ala Leu Ser Lys
435 440 445
Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Val Thr Ala
450 455 460

Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys Glx
465 470 475
<210> 55
<211> 607
<212> PRT
<213> Artificial Sequence
<220>
<223> Description of Artificial Sequence: synthetic
<400> 55
Gly Ser Ala Thr Ala Ala Thr Val Pro Pro Ala Ala Pro Ala Gly Glu
1 5 10 15
Gly Gly Pro Pro Ala Pro Pro Pro Asn Leu Thr Ser Asn Arg Arg Leu
20 25 30
Gln Gln Thr Gln Ala Gln Val Glu Asp Glu Val Val Asp Ile Met Arg
35 40 45

Val Asn Val Asp Lys Val Leu Glu Arg Asp Gin Lys Leu Ser Glu Leu
50 55 60
19


CA 02325597 2001-03-21

Asp Asp Arg Ala Asp Ala Leu Gln Ala Gly Ala Ser Gln Phe Glu Thr
65 70 75 80
Ser Ala Ala Lys Leu Lys Arg Lys Tyr Trp Trp Lys Asn Leu Lys Met
85 90 95

Met Ile Leu Leu Gly Val Ile Cys Ala Ile Ile Leu Val Ile Ile Ile
100 105 110
Val Tyr Phe Ser Thr Gly Ser Gly Ser Gly Ser Gly Ser Gly Ser Gly
115 120 125
Pro Val Ser Lys Gly Glu Glu Leu Phe Thr Gly Val Val Pro Ile Leu
130 135 140

Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser Gly
145 150 155 160
Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Phe Ile
165 170 175

Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr Thr
180 185 190
Leu Thr His Gly Val Gln Cys Phe Ser Arg Tyr Pro Asp His Met Lys
195 200 205
Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln Glu
210 215 220
Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala Glu
225 230 235 240
Val Lys Phe Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Gly
245 250 255

Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Giu Tyr
260 265 270
Asn Phe Asn Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn
275 280 285
Gly Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser
290 295 300

Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly
305 310 315 320
Pro Val Leu Leu Pro Asp Asn His Tyr Leu Ser Thr Gln Ser Ala Leu
325 330 335

Ser Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe
340 345 350
Val Thr Ala Ala Gly Ser Gly Ser Ile Glu Gly Arg Arg Lys Leu Gln
355 360 365
Gly Ser Gly Ser Lys Gly Glu Glu Leu Thr Phe Gly Val Val Pro Ile
370 375 380
Leu Val Glu Leu Asp Gly Asp Val Asn Gly His Lys Phe Ser Val Ser
385 390 395 400


CA 02325597 2001-03-21

Gly Glu Gly Glu Gly Asp Ala Thr Tyr Gly Lys Leu Thr Leu Lys Ile
405 410 415
Phe Cys Thr Thr Gly Lys Leu Pro Val Pro Trp Pro Thr Leu Val Thr
420 425 430
Thr Leu Thr Tyr Gly Val Gln Cys Phe Ser Arg Tyr Pro Asp His Met
435 440 445

Lys Gln His Asp Phe Phe Lys Ser Ala Met Pro Glu Gly Tyr Val Gln
450 455 460
Glu Arg Thr Ile Phe Phe Lys Asp Asp Gly Asn Tyr Lys Thr Arg Ala
465 470 475 480
Glu Val Lys Glu Gly Asp Thr Leu Val Asn Arg Ile Glu Leu Lys Giy
485 490 495
Ile Asp Phe Lys Glu Asp Gly Asn Ile Leu Gly His Lys Leu Glu Tyr
500 505 510

Asn Tyr Asn Ser His Asn Val Tyr Ile Met Ala Asp Lys Gln Lys Asn
515 520 525
Gly Ile Lys Val Asn Phe Lys Ile Arg His Asn Ile Glu Asp Gly Ser
530 535 540
Val Gln Leu Ala Asp His Tyr Gln Gln Asn Thr Pro Ile Gly Asp Gly
545 550 555 560
Pro Val Leu Leu Pro Asn His Tyr Leu Ser Thr Gln Ser Ala Leu Ser
565 570 575
Lys Asp Pro Asn Glu Lys Arg Asp His Met Val Leu Leu Glu Phe Val
580 585 590
Thr Ala Ala Gly Ile Thr Leu Gly Met Asp Glu Leu Tyr Lys Glx
595 600 605

21

Representative Drawing

Sorry, the representative drawing for patent document number 2325597 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2010-09-21
(86) PCT Filing Date 1999-04-16
(87) PCT Publication Date 1999-10-28
(85) National Entry 2000-10-16
Examination Requested 2004-04-13
(45) Issued 2010-09-21
Expired 2019-04-16

Abandonment History

There is no abandonment history.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $300.00 2000-10-16
Registration of a document - section 124 $100.00 2001-03-21
Registration of a document - section 124 $100.00 2001-03-21
Maintenance Fee - Application - New Act 2 2001-04-17 $100.00 2001-04-04
Maintenance Fee - Application - New Act 3 2002-04-16 $100.00 2002-04-03
Maintenance Fee - Application - New Act 4 2003-04-16 $100.00 2003-04-16
Maintenance Fee - Application - New Act 5 2004-04-16 $200.00 2004-02-26
Request for Examination $800.00 2004-04-13
Maintenance Fee - Application - New Act 6 2005-04-18 $200.00 2005-03-14
Maintenance Fee - Application - New Act 7 2006-04-18 $200.00 2006-04-05
Maintenance Fee - Application - New Act 8 2007-04-16 $200.00 2007-04-02
Maintenance Fee - Application - New Act 9 2008-04-16 $200.00 2008-04-01
Maintenance Fee - Application - New Act 10 2009-04-16 $250.00 2009-03-31
Maintenance Fee - Application - New Act 11 2010-04-16 $250.00 2010-03-31
Final Fee $300.00 2010-07-02
Maintenance Fee - Patent - New Act 12 2011-04-18 $250.00 2011-03-30
Maintenance Fee - Patent - New Act 13 2012-04-16 $250.00 2012-03-30
Maintenance Fee - Patent - New Act 14 2013-04-16 $250.00 2013-04-01
Maintenance Fee - Patent - New Act 15 2014-04-16 $450.00 2014-04-14
Maintenance Fee - Patent - New Act 16 2015-04-16 $450.00 2015-04-13
Maintenance Fee - Patent - New Act 17 2016-04-18 $450.00 2016-04-11
Maintenance Fee - Patent - New Act 18 2017-04-18 $450.00 2017-04-10
Maintenance Fee - Patent - New Act 19 2018-04-16 $450.00 2018-04-09
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
RIGEL PHARMACEUTICALS, INC.
Past Owners on Record
FISHER, JOSEPH
LORENS, JAMES
PAYAN, DONALD
ROSSI, ALEXANDER
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2008-09-10 78 3,854
Claims 2008-09-10 2 60
Drawings 2008-09-10 10 291
Description 2000-10-16 55 3,394
Description 2001-03-21 76 3,830
Abstract 2000-10-16 1 46
Claims 2000-10-16 2 44
Drawings 2000-10-16 10 287
Cover Page 2001-01-16 1 34
Cover Page 2010-08-25 1 33
Prosecution-Amendment 2006-04-19 1 42
Correspondence 2000-12-28 1 41
Assignment 2000-10-16 3 91
PCT 2000-10-16 9 320
Prosecution-Amendment 2000-12-21 1 45
Correspondence 2001-01-23 2 122
Correspondence 2001-03-21 23 492
Assignment 2001-03-21 16 614
Prosecution-Amendment 2004-04-13 1 38
Prosecution-Amendment 2007-01-31 1 43
Prosecution-Amendment 2007-05-03 1 41
Prosecution-Amendment 2007-08-27 1 37
Prosecution-Amendment 2007-10-17 1 37
Prosecution-Amendment 2008-03-12 5 230
Prosecution-Amendment 2008-09-10 34 1,735
Correspondence 2010-07-02 1 37

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :